U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

National Clinical Guideline Centre (UK). Psoriasis: Assessment and Management of Psoriasis. London: Royal College of Physicians (UK); 2012 Oct. (NICE Clinical Guidelines, No. 153.)

  • Update information September 2017: The guideline has been revised throughout to link to MHRA advice and NICE technology appraisals that have been completed since original publication. Minor updates since publication August 2019: Links to the MHRA safety advice on the risk of using retinoids in pregnancy have been updated to the June 2019 version.

Update information September 2017: The guideline has been revised throughout to link to MHRA advice and NICE technology appraisals that have been completed since original publication. Minor updates since publication August 2019: Links to the MHRA safety advice on the risk of using retinoids in pregnancy have been updated to the June 2019 version.

Cover of Psoriasis

Psoriasis: Assessment and Management of Psoriasis.

Show details

8Topical therapy

Topical therapy in some form or another is prescribed to virtually everyone with psoriasis presenting for treatment. The majority of people with psoriasis have localised disease and here, topical therapy is the principal approach to treatment. In more extensive and severe forms of psoriasis, topical therapy remains an important adjunct to second and third line therapy and remains the mainstay of treatment in people who do not want or cannot use second or third line therapies.

Corticosteroids, vitamin D3 and its analogues, calcineurin inhibitors, retinoids, tar, dithranol and keratolytic agents such as salicylic acid and urea are available for topical use for psoriasis and come in a vast array of different formulations, combinations, potencies and dilutions. Some of the topical agents in common use - particularly in specialist settings - are ‘special manufacture’ medicines (‘Specials’)44. Preparations such as dithranol in Lassar’s paste and crude coal tar are sometimes referred to as ‘complex topicals’ as they usually needs to be administered in specialist settings by trained individuals to optimise outcomes and minimise adverse effects including irritation and staining of skin.

For most patients, topical treatments are prescribed for home use to self-manage psoriasis. Variable outcomes are reported with the use of topical therapies and much of this variation is likely to relate to problems with adherence. Adherence, previously referred to as compliance, is the degree to which a patients’ behaviour taking or using treatments corresponds with recommendations from a healthcare professional. Adherence can be sub-divided into primary adherence, which is redemption of prescriptions and secondary adherence, which relates to correct use of treatments. Primary adherence in one study was found to be low with 30% of patients not collecting their prescriptions393. This study also revealed that 95% of patients under-dosed with their topical treatment. Moreover, secondary adherence to topical therapies is variable with one study showing that 39% of patients did not adhere to the recommended treatment regime331 while another reported a mean adherence of 72%434. There are several factors that influence secondary adherence such as the cosmetic acceptability of the product, time required for application, dosage regimes as well as ease of use. The cosmetic acceptability of a product is related to the formulation and can have an impact on secondary adherence. In one survey of psoriasis patients prescribed topical therapies it was found that the greasiness of the preparation was responsible for non-adherence in 11% of patients409. Ointments have been traditionally used due to perceived superior efficacy and the fact that the vehicle is more effective at hydrating dry, scaling psoriatic skin. However, some evidence suggests that patients prefer a cream or gel formulation153 and potential differences in vehicles may have a negative impact on adherence and should be discussed with patients when prescribing topical agents.

Although several factors influence adherence, one suggested technique to improve adherence is through patient education. In a recent focus group study with psoriatic patients, it was noted how patients identified that instruction on the correct use topical treatments was essential but often absent from consultations. The study also revealed the erratic and inconsistent use of topical treatments by patients, therefore highlighting the need for more effective community-based support89. There is some evidence that adjunctive patient education improves both quality of life and reduces disease severity in patients with skin disease71 and this approach has been successfully deployed in studies with psoriatic patients3,361.

Health professional prescribing topical therapies should have sufficient product knowledge including the effect of the treatment on psoriatic plaques and any adverse effects on the surrounding skin. Prescribers also need to engage with patients in an attempt to ascertain the psychological impact of their psoriasis and to agree therapeutic goals in an effort to improve adherence. Support for patients with dexterity or disability problems can be provided together with advice to patients to support adherence. In addition, the medicines use review service may provide information about usage of treatments and where necessary, provide knowledge to help to resolve poor or ineffective use of therapies.

The wide array of potential topical agents available requires that healthcare professionals treating psoriasis deploy a therapeutic strategy that is based on the best available evidence. Such an approach is justified, not only to endeavour to provide a high standard of care but to ensure that referrals to specialist centres are appropriately managed. In an effort to provide health professionals with an algorithm for sequencing of topical agents and for criteria that would trigger a referral, we examined the evidence to determine the most suitable strategic approach for the individual patient.

There is a general consensus amongst clinicians and patients that emollients are useful adjunctive therapy in the management of inflammatory skin disease including psoriasis. Emollients help to restore pliability to the skin and can improve the cosmetic appearance of plaques by reducing shedding of scale. Emollients also appear to reduce pruritus and can help to reduce cracking of the skin which can be extremely painful. The GDG felt that the use of emollients in psoriasis was widespread and of accepted value, and review of the evidence was unlikely to yield important data that would justify recommending a change in practice. We have therefore limited our evidence review to active topical therapies in psoriasis. We have also focussed our review on plaque psoriasis only for pragmatic reasons, given the number of studies in this area, but acknowledge that topical therapies are also key components of treatment for other types of psoriasis.

The face, flexures (including genitals) and scalp are often described as 'difficult to treat' since the face and flexures are especially vulnerable to tolerability and toxicity issues, and the scalp is difficult to access and often resistant to treatment. These sites are also often 'high impact' sites, and in one recent patient survey318 the number of people with scalp psoriasis was notable (1158 out of 1618 respondents reported having scalp psoriasis) and clearance of visible areas was rated as important. The GDG therefore felt these sites should be given special consideration when considering the evidence. The GDG were also interested to establish the timelines for treatment response of the various agents to guide clinicians on when to review patients in order to optimise outcomes, and limit use of ineffective agents. The GDG posed the following questions:

In people with chronic plaque psoriasis of the trunk and/or limbs, (i), what are the clinical effectiveness, safety, tolerability, and cost effectiveness of topical vitamin D and vitamin D analogues, potent or very potent corticosteroids, tar, dithranol and retinoids compared with placebo or vitamin D and vitamin D analogues, and of combined or concurrent vitamin D and vitamin D analogues and potent corticosteroids compared with potent corticosteroid or vitamin D and vitamin D analogues alone??; and (ii) at what time interval should the patient be reviewed to assess the effectiveness of treatment with topical therapy?

In people with psoriasis at difficult-to-treat sites (scalp, flexures including genitals, face), (i)what are the clinical effectiveness, safety, tolerability and cost effectiveness of vitamin D and vitamin D analogues, mild to very potent corticosteroids, combined or concurrent vitamin D or vitamin D analogue and potent corticosteroid, pimecrolimus, tacrolimus, tar, dithranol and retinoids compared with placebo, corticosteroids or vitamin D or vitamin D analogues?; and (ii) at what time interval should the patient be reviewed to assess the effectiveness of treatment with topical therapy?

8.1. Topical therapies for trunk and limb psoriasis

8.1.1. Methodological introduction

A literature search was conducted for RCTs or systematic reviews that addressed the efficacy and safety of topical vitamin D and vitamin D analogues, potent or very potent corticosteroids, combined vitamin D or vitamin D analogue and potent corticosteroid, concurrent vitamin D or vitamin D analogue and potent corticosteroid (one applied in the morning and one in the evening) tar, dithranol and retinoids for induction or maintenance of remission in people with psoriasis. No time limit was placed on the literature search and there were no limitations duration of follow-up. However, the sample size had to be at least 25 participants per study arm and indirect populations were excluded.

The evidence considered included topical monotherapies compared with vitamin D or vitamin D analogue or with placebo/vehicle, while combined or concurrent vitamin D or vitamin D analogue and potent corticosteroid were compared with the constituent monotherapies (and not with placebo). Studies only comparing different dosages or formulations of the same intervention were excluded. Similarly, studies comparing interventions within the classes of either vitamin D and its analogues or corticosteroids were excluded (unless the comparison pertained to frequency of administration e.g., once or twice daily dosing). A class effect was assumed for these agents and so data on all vitamin D and its analogues was pooled into one analysis as was data on any potent corticosteroids and on very potent corticosteroids, unless heterogeneity was found.

The outcomes considered were:

  • Clear/nearly clear or marked improvement (at least 75% improvement) on Investigator’s assessment of overall global improvement (IAGI) or clear/nearly clear/minimal (not mild) on Physician’s Global Assessment (PGA)
  • Clear/nearly clear or marked improvement (at least 75% improvement) on Patient’s assessment of overall global improvement (PAGI) or clear/nearly clear/minimal (not mild) on Patient’s Global Assessment
  • Percentage change in PASI – change is represented by a negative value if the PASI score decreased
  • Change in DLQI
  • Duration of remission
  • Time-to-remission or time-to-maximum effect based on IAGI, PGA, PASI or total severity score (to address part ii of the question)*
  • Withdrawal due to toxicity
  • Withdrawal due to lack of efficacy
  • Skin atrophy

A published Cochrane Review238 was identified from the literature search, which at the time of development of this guideline was being updated and publication of which would not fall within the development period of this guideline. However, the original Cochrane Review was not able to be updated directly owing to differences in methodology and in outcomes, which did not match those required to feed into a novel health economics model. The Cochrane reference list and literature search protocols were used for cross-referencing and their published literature search was re-run to update it. Additionally, following close collaboration and discussion with the Cochrane Skin Group, study characteristic and withdrawal outcome data were extracted directly from the published Cochrane Review to enable novel meta analysis.

In addition to the Cochrane Review, 54 RCTs were found that addressed the question and were included in the review28,15,25,29,32,47,58,81,125,141,148,178,198 49,66,73,79,102,132,156,167,174,179,195197,201,208,210,211,216,227,246,251,255,295,298,302,311,313,344,346,351,354,360,381,400,401,410,411,417420,428. However, just two studies178,195,196 directly assessed maintenance treatment and just one study was conducted in a paediatric population295.

The included studies differed in terms of the disease severity and treatment duration (Table 61). Note the potential limitation of studies comparing interventions that act over different periods (e.g., the faster acting clobetasol propionate and the slower acting calcipotriol), especially if the treatment duration chosen for the trial does not permit the maximum effect of the slower acting intervention to be observed.

Table 61. Characteristics of included studies.

Table 61

Characteristics of included studies.

Data from within-patient trials should be adjusted for the correlation coefficient relating to the comparison of paired data. None of the included studies reported this statistic; neither did they report sufficient detail for it to be calculated. Where possible, within- and between-patient data were pooled, accepting that this may result in underweighting of the within-patient studies. This is a conservative estimate. Sensitivity analyses were undertaken to investigate whether the effect size varied consistently for within- and between-patient studies. There was no evidence that the size of effect varied in a systematic way and it was often not possible to say if consistent differences were present as there was only one within patient study for a given comparison.

8.1.2. Vitamin D and vitamin D analogue vs. placebo

8.1.2.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D and vitamin D analoguesplaceboRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) - Calcipotriol OD (follow-up 4–8 weeks)
3
Barker1999
Fleming2010A
Kaufmann2002
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionNone129/587 (22%)17/223 (7.6%)RR 2.78 (1.75 to 4.41)136 more per 1000 (from 57 more to 260 more)⊕⊕⊕○
MODERATE
Investigator’s assessment (clear/nearly clear) - Calcipotriol BD (follow-up 4–8 weeks)
4
Dubertret 1992
Guenther 2002
Highton 1995
Papp 2003
randomised trialsseriousbno serious inconsistencyno serious indirectnessno serious imprecisionNone351/721 (48.7%)61/498 (12.2%)RR 4.48 (3.5 to 5.73)426 more per 1000 (from 306 more to 579 more)⊕⊕⊕○
MODERATE
Investigator’s assessment (clear/nearly clear) - Calcitriol OD (follow-up 10 weeks)
1
Perez 1996
randomised trialsseriouscno serious inconsistencyno serious indirectnessdno serious imprecisionNone37/84 (44%)0/84 (0%)RR 75 (4.68 to 1201.67)-⊕⊕⊕○
MODERATE
Investigator’s assessment (clear/nearly clear) - Calcitriol BD (follow-up 6 weeks)
2
Langner 1992
Langner 1993
randomised trialsseriouseno serious inconsistencyseriousfno serious imprecisionNone45/61 (73.8%)22/61 (36.1%)RR 2.05 (1.42 to 2.95)379 more per 1000 (from 151 more to 703 more)⊕⊕○○
LOW
Investigator’s assessment (clear/nearly clear) - Tacalcitol (OD) (follow-up 8 weeks)
1
Langley 2011A
randomised trialsvery seriousgno serious inconsistencyno serious indirectnessno serious imprecisionNone33/184 (17.9%)5/91 (5.5%)RR 3.26 (1.32 to 8.08)124 more per 1000 (from 18 more to 389 more)⊕⊕○○
LOW
Patient’s assessment (clear/nearly clear) - Calcipotriol OD or BD (follow-up 4–8 weeks)
3
Kaufmann 2002
Guenther 2002
Harrington 1996
randomised trialsserioushno serious inconsistencyno serious indirectnessno serious imprecisionNone402/988 (40.7%)54/434 (12.4%)RR 3.35 (2.58 to 4.34)292 more per 1000 (from 197 more to 416 more)⊕⊕⊕○
MODERATE
Patient’s assessment (clear/nearly clear) - Tacalcitol (OD) (follow-up 8 weeks)
1
Langley 2011A
randomised trialsvery seriousino serious inconsistencyno serious indirectnessvery seriousjNone35/163 (21.5%)14/64 (21.9%)RR 0.98 (0.57 to 1.7)4 fewer per 1000 (from 94 fewer to 153 more)⊕○○○
VERY LOW
% change in PASI - Calcipotriol BD (follow-up 4 weeks) (Better indicated by lower values)
1
Dubertret 1992
randomised trialsseriouscno serious inconsistencyno serious indirectnessno serious imprecisionNone6060-MD 23.2 lower (35.57 to 10.83 lower)⊕⊕⊕○
MODERATE
Withdrawals due to adverse events – Calcipotriol, calcitriol or tacalcitol OD or BD (follow-up 4–8 weeks)
11
Barker 1999
Kaufmann 2002
Guenther 2002
Harrington 1996
Highton 1995
Langner 1992
Langner 1993
Langley 2011A
Perez 1996
Scarpa 1997
van de Kerkhof 1996
randomised trialskseriouslno serious inconsistencyno serious indirectnessmseriousnData40/1736 (2.3%)31/1055 (2.9%)RR 0.62 (0.4 to 0.97)11 fewer per 1000 (from 1 fewer to 18 fewer)⊕⊕○○
LOW
Withdrawals due to lack of efficacy – Calcipotriol or calcitriol OD or BD (follow-up 4–8 weeks)
7
Barker 1999
Guenther 2002
Harrington 1996
Langner 1992
Langner 1993
Perez 1996
Scarpa 1997
randomised trialsoseriouspno serious inconsistencyno serious indirectnessno serious imprecisionNone3/893 (0.34%)22/644 (3.4%)RR 0.15 (0.05 to 0.42)29 fewer per 1000 (from 20 fewer to 32 fewer)⊕⊕⊕○
MODERATE
Skin atrophy – Calcipotriol BD (follow-up 4 weeks)
2
Guenther 2002
Papp 2003
randomised trialsseriousqno serious inconsistencyno serious indirectnessvery seriousjNone1/535 (0.19%)1/316 (0.32%)RR 0.92 (0.06 to 14.56)0 fewer per 1000 (from 3 fewer to 43 more)⊕○○○
VERY LOW
Relapse rate at 8 weeks post-treatment - Tacalcitol OD (follow-up 8 weeks)
1
Langley 2011A
randomised trialsvery seriousrno serious inconsistencyserioussseriousnNone7/31 (22.6%)3/5 (60%)RR 0.38 (0.14 to 0.99)372 fewer per 1000 (from 6 fewer to 516 fewer)⊕○○○
VERY LOW
Median time to relapse - Tacalcitol OD (follow-up 8 weeks post treatment)
1
Langley 2011A
randomised trialsvery seriousrno serious inconsistencyno serious indirectnessserioustNone315-61 days in both groups⊕○○○
VERY LOW
a

3/3 unclear allocation concealment; 1/3 (93.4% weighted) differential dropout (8.1%: calcipotriol; 15.9%: vehicle); 1/3 (4% weighted) baseline clinical characteristics not reported

b

4/4 unclear allocation concealment; 2/4 unclear blinding; 1/4 (35% weighted) unclear if dropout rate was evenly distributed between study arms

c

Unclear allocation concealment and blinding

d

Study used Vaseline as the placebo (not vehicle)

e

2/2 unclear allocation concealment and blinding; 1/2 studies (40.9% weighted) treatment stopped if at least one side cleared; therefore, lesion on contra lateral side may have clear if treated for the full study period

f

1/2 studies used high concentration of calcitriol (15 μg/g, licensed at 3 μg/g)

g

Unclear allocation concealment and blinding; high differential dropout rate: 11.4% tacalcitol; 29.7% placebo

h

3/3 unclear allocation concealment; 2/3 studies (61.4% weighted) higher but acceptable dropout in vehicle group

i

Unclear allocation concealment and single blinded (investigator); high dropout rate in placebo group (tacalcitol: 11.4%; placebo: 29.7%

j

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

k

For 3/9 (Barker, Scarpa and van de Kerkhof) studies data were taken from a published Cochrane Review

l

10/11 unclear allocation concealment; 3/11 unclear blinding (20.6% weighted); 3/11 higher dropout rate in placebo group; 1/11 (3.4% weighted) unclear baseline clinical characteristics

m

In one study (weighted 1.1%) 24.6% of patients test lesions were localised on the face or face and other parts of the body; one study used a very high concentration of calcitriol (weighted 1.1%)

n

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit to no clinically important benefit)

o

For 1/4 studies (Barker) data were taken from a published Cochrane Review

p

7/7 unclear allocation concealment; 1/7 (6.1% weighted) unclear baseline clinical characteristics; 1/7 (9.7% weighted) higher dropout in placebo group

q

2/2 unclear allocation concealment

r

Unclear allocation concealment and single blinded (investigator); high dropout rate in placebo group (tacalcitol: 11.4%; placebo: 29.7%); also, unclear baseline comparability as only includes those in each group who achieved remission; therefore, there are fewer participants in the placebo group

s

Surrogate outcome for duration of remission

t

No range provided

8.1.2.2. Evidence statements

In people with psoriasis, topical vitamin D or vitamin D analogue treatment was statistically significantly better than placebo for:

  • Investigator’s assessment (clear/nearly clear on PGA) at 4–10 weeks for calcipotriol once daily, calcipotriol twice daily, calcitriol once daily, calcitriol twice daily or tacalcitol once daily [11 studies (7 between- and 4 within-patient studies); 2387 participants (2594 randomised units); low to moderate quality evidence]25,81,102,132,148,179,208,210,211,302,311
  • Patient assessment (clear/nearly clear on PGA) at 4–8 weeks for calcipotriol once daily or calcipotriol twice daily [3 between-patient studies; 1432 participants; moderate quality evidence]132,141,179
  • Percentage change in PASI at 4 weeks for calcipotriol twice daily [1 within-patient study; 60 participants (120 randomised units); moderate quality evidence]81
  • Withdrawal due to adverse events at 4–8 weeks [11 studies (6 between- and 5 within-patient); 2367 participants (2791 randomised units); low quality evidence] 25,132,141,148,179,208,210,211,311,354
  • Withdrawal due to lack of efficacy at 4–8 weeks [7 studies (4 between- and 3 within-patient); 1207 participants (1477 randomised units); moderate quality evidence]25,132,141,210,211,311,354
  • Relapse at 8 weeks post treatment with tacalcitol once daily [1 between-patient study; 36 participants; very low quality evidence]208.

In people with psoriasis, there was no statistically significant difference between topical vitamin D or vitamin D analogue treatment and placebo for:

  • Patient assessment at 8 weeks (clear/nearly clear) with tacalcitol once daily [1 between-patient study; 227 participants; very low quality evidence]208
  • Skin atrophy at 4 weeks for calcipotriol twice daily [2 between-patient studies; 851 participants; very low quality evidence]132,302
Evidence statement for individual study where no statistical analysis could be performed

In people with psoriasis, there was no difference between topical vitamin D or vitamin D analogue treatment and placebo for:

  • Median time-to-relapse among those who had achieved remission with tacalcitol once daily (followed for up to 8 weeks post treatment) [1 study; 36 participants; very low quality evidence]208.

8.1.2.3. Heterogeneity

  • There was significant heterogeneity between data regarding the investigator’s assessment of efficacy. This heterogeneity was removed by creating subgroups based on the specific agent and treatment frequency of the vitamin D or vitamin D analogue. Nevertheless, all agents and frequencies demonstrated a clinically significant benefit compared with placebo.
  • There was significant heterogeneity between data regarding the patient’s assessment of efficacy. This heterogeneity was removed by creating subgroups based on the specific agent within the vitamin D or vitamin D analogue class, while treatment frequency did not explain the differences. It appeared that tacalcitol was not more effective than placebo based on patient’s assessment, whereas calcipotriol was more effective. However, the heterogeneity may also have been caused by the tacalcitol study having a higher risk of bias as it was only investigator blinded (although this may be more likely to increase the effect estimate in favour of the active intervention) and had a 30% drop-out rate in the placebo group.
  • There was no significant heterogeneity for the remaining outcomes

8.1.3. Vitamin D or vitamin D analogue vs. placebo (children)

8.1.3.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D or vitamin D analoguesplaceboRelative
(95% CI)
Absolute
Investigator’s assessment (clear/nearly clear) - Calcipotriol BD (follow-up 8 weeks)
1
Oranje 1997
randomised trialsseriousano serious inconsistencyno serious indirectnessseriousbnone26/43 (60.5%)15/34 (44.1%)RR 1.37 (0.87 to 2.15)163 more per 1000 (from 57 fewer to 507 more)⊕⊕○○
LOW
Patient’s assessment (clear/nearly clear) - Calcipotriol BD (follow-up 8 weeks)
1
Oranje 1997
randomised trialsseriousano serious inconsistencyno serious indirectnessvery seriouscnone21/43 (48.8%)16/34 (47.1%)RR 1.04 (0.65 to 1.66)19 more per 1000 (from 165 fewer to 311 more)⊕○○○
VERY LOW
% change in PASI - Calcipotriol BD (follow-up 8 weeks)
1
Oranje 1997
randomised trialsseriousano serious inconsistencyno serious indirectnessseriousbnone4334-MD 14.90 lower (34.69 lower to 4.89 higher)⊕⊕○○
LOW
a

Unclear allocation concealment and blinding; acceptable drop-out rates but higher with calcipotriol

b

Confidence interval ranges from clinically significant effect to no effect

c

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

8.1.3.2. Evidence statements

In children with psoriasis, there was no statistically significant difference between calcipotriol twice daily and placebo for:

  • Investigator’s assessment (clear/nearly clear) at 8 weeks [1 between-patient study; 77 participants; low quality evidence]295
  • Patients assessment (clear/nearly clear) at 8 weeks [1 between-patient study; 77 participants; very low quality evidence295
  • % change in PASI at 8 weeks [1 between-patient study; 77 participants; low quality evidence295

8.1.3.3. Heterogeneity

  • Not applicable as only one study assessed vitamin D or vitamin D analogues compared with placebo in children

8.1.4. Potent corticosteroid vs. placebo

8.1.4.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCorticosteroid (potent)PlaceboRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) – Mometasone furoate OD, hydrocortisone butyrate BD, betamethasone dipropionate OD or BD (follow-up 3–8 weeks)
6
Fleming2010A
Kaufmann 2002
Papp 2003
Wortzel 1975
Medansky 1987
Sears 1997
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionNone409/1038 (39.4%)36/469 (7.7%)RR 4.68 (3.38 to 6.48)282 more per 1000 (from 183 more to 421 more)⊕⊕⊕○
MODERATE
Patient’s assessment (clear/nearly clear) – hydrocortisone butyrate BD or betamethasone dipropionate OD (follow-up 3–4 weeks)
2
Kaufmann 2002
Sears 1997
randomised trialsseriousbno serious inconsistencyno serious indirectnessno serious imprecisionNone228/554 (41.2%)17/240 (7.1%)RR 4.88 (3.06 to 7.77)275 more per 1000 (from 146 more to 480 more)⊕⊕⊕○
MODERATE
Withdrawals due to adverse events - Once daily potent corticosteroid (mometasone furoate or betamethasone dipropionate) (follow-up 3–4 weeks)
2
Kaufmann 2002
Medansky 1987
randomised trialsseriouscno serious inconsistencyno serious indirectnessno serious imprecisionNone5/502 (1%)15/191 (7.9%)RR 0.13 (0.05 to 0.36)68 fewer per 1000 (from 50 fewer to 75 fewer)⊕⊕⊕○
MODERATE
Withdrawals due to adverse events - Twice daily potent corticosteroid (hydrocortisone butyrate, betamethasone valerate or betamethasone dipropionate) (follow-up 3–12 weeks)
3
Sears 1997
Stein 2001
Wortzel 1975
randomised trialsseriousdno serious inconsistencyno serious indirectnessevery seriousfNone4/163 (2.5%)0/162 (0%)RR 5.02 (0.6 to 42.26)-⊕○○○
VERY LOW
Withdrawals due to lack of efficacy - Betamethasone dipropionate BD (follow-up 3 weeks)
1
Wortzel 1975
randomised trialsno serious risk of biasno serious inconsistencyno serious indirectnessno serious imprecisionNone0/39 (0%)0/37 (0%)not poolednot pooled⊕⊕⊕⊕
HIGH
Skin atrophy – Mometasone furoate OD or betamethasone dipropionate BD (follow-up 3–4 weeks)
2
Papp 2003
Medansky 1987
randomised trialsseriousgno serious inconsistencyno serious indirectnessvery seriousfNone2/363 (0.55%)0/153 (0%)RR 1.74 (0.08 to 35.87)-⊕○○○
VERY LOW
a

5/6 unclear allocation concealment; 2/6 unclear blinding; 1/6 high dropout rate (weighted 15%); 1/6 (49% weighted) differential dropout rate: 4.6% betamethasone, 15.9% placebo

b

Unclear allocation concealment and blinding

c

2/2 unclear allocation concealment; 1/2 unclear blinding; 1/2 (16.5% weighted) high dropout rate (21.5% from steroid and 26.3% from placebo)

d

1/3 inadequate and 1/3 unclear allocation concealment; 2/3 unclear blinding

e

Data for Stein study taken from published Cochrane Review

f

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

g

2/2 unclear allocation concealment; 1/2 (0% weighted) unclear blinding and high dropout rate (21.5% corticosteroids and 26.3% placebo)

8.1.4.2. Evidence statements

In people with psoriasis, topical potent corticosteroid treatment was statistically significantly better than placebo for:

  • Investigator’s assessment (clear/nearly clear) at 3–8 weeks for mometasone furoate once daily, hydrocortisone butyrate twice daily and betamethasone dipropionate once or twice daily [6 between-patient studies; 1507 participants; moderate quality evidence]102,179,246,302,360,428
  • Patient’s assessment (clear/nearly clear) at 3–4 weeks for hydrocortisone butyrate twice daily or betamethasone dipropionate once daily [2 between-patient studies; 794 participants; moderate quality evidence]179,360
  • Withdrawal due to adverse events at 3–4 weeks for potent corticosteroid (mometasone furoate or betamethasone dipropionate) once daily [2 between-patient studies; 693 participants; moderate quality evidence]179,246

In people with psoriasis, there were no events with either topical potent corticosteroid treatment or placebo for:

  • Withdrawal due to lack of efficacy at 3 weeks for betamethasone dipropionate twice daily [1 between-patient study; 76 participants; high quality evidence]428

In people with psoriasis, there was no statistically significant difference between topical potent corticosteroid treatment and placebo for:

  • Withdrawal due to adverse events at 3–12 weeks for potent corticosteroid (hydrocortisone butyrate, betamethasone valerate or betamethasone dipropionate) twice daily [3 studies (2 between- and 1 within-patient); 285 participants (325 randomised units); very low quality evidence]360,381,428
  • Skin atrophy [2 between-patient studies; 516 participants; very low quality evidence]246,302

8.1.4.3. Heterogeneity

  • There was significant heterogeneity between data regarding withdrawals due to adverse effects. This heterogeneity was removed by creating subgroups based on treatment frequency. It was considered clinically more likely that the treatment frequency was causing the heterogeneity rather than the specific agent within the potent corticosteroid class.
  • There was no significant heterogeneity for the remaining outcomes

8.1.5. Very potent corticosteroid vs. placebo

8.1.5.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCorticosteroid (very potent)placeboRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) – clobetasol propionate OD or BD (follow-up 2–4 weeks)
5
Decroix 2004
Gottlieb 2003C
Jarratt 2006
Lebwohl 2002
Lowe 2005
randomised trialsvery seriousaseriousbno serious indirectnessno serious imprecisionNone370/592 (62.5%)35/267 (13.1%)RR 6.45 (2.63 to 15.81)714 more per 1000 (from 214 more to 1000 more)⊕○○○
VERY LOW
Patient’s assessment (clear/nearly clear) - Clobetasol propionate BD (follow-up 2 weeks)
2
Gottlieb 2003C
Lebwohl 2002
randomised trialsvery seriouscno serious inconsistencyno serious indirectnessno serious imprecisionNone87/200 (43.5%)37/160 (23.1%)RR 2.23 (1.62 to 3.05)284 more per 1000 (from 143 more to 474 more)⊕⊕○○
LOW
Withdrawals due to adverse events – clobetasol propionate OD or BD (follow-up 2–4 weeks)
7
Beutner 2006
Decroix 2004
Gottlieb 2003C
Jarratt 2006
Jorizzo 1997
Lebwohl 2002
Lowe 2005
randomised trialsvery seriousdno serious inconsistencyno serious indirectnessvery seriouseNone3/653 (0.46%)2/331 (0.60%)RR 0.56 (0.12 to 2.52)4 fewer per 1000 (from 8 fewer to 13 more)⊕○○○
VERY LOW
Withdrawals due to lack of efficacy - Clobetasol propionate OD or BD (follow-up 4 weeks)
3
Decroix 2004
Beutner 2006
Jarratt 2006
randomised trialsseriousfno serious inconsistencyno serious indirectnessvery seriouseNone0/268 (0%)1/117 (0.85%)RR 0.06 (0 to 1.44)8 fewer per 1000 (from 5 fewer to 9 more)⊕○○○
VERY LOW
Skin atrophy - Clobetasol propionate OD or BD (follow-up 4 weeks)
4
Beutner 2006
Decroix 2004
Jarratt 2006
Jorizzo 1997
randomised trialsseriousgno serious inconsistencyno serious indirectnessvery seriouseNone7/308 (2.3%)0/156 (0%)RR 2.7 (0.16 to 46.15)-⊕○○○
VERY LOW
a

5/5 unclear allocation concealment; 3/5 unclear blinding; 2/5 single blind (investigator); 1/5 (2.1% weighted) high dropout rate: 27.6% in placebo group, 6.1% and 4.9% in clobetasol lotion and cream; 1/5 (67.3% weighted) unclear baseline demographics; 1/5 (21.7% weighted) fewer males in clobetasol group

b

Heterogeneity was present (I2 = 70%) that could not be explained by pre-defined subgroups (however, all studies showed the same direction of effect)

c

2/2 unclear allocation concealment and blinding; 1/2 (96% weighted) unclear baseline demographics

d

7/7 unclear allocation concealment; 5/7 unclear blinding and 2/7 single blinded (investigator); 1/7 (35.6% weighted) unclear baseline demographics; 2/7 (44% weighted) high differential dropout rate

e

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

f

3/3 unclear allocation concealment; 2/3 unclear blinding and 1/3 single blind (investigato

g

4/4 unclear allocation concealment; 3/4 unclear blinding and 1/4 single blind (investigator)

8.1.5.2. Evidence statements

In people with psoriasis, topical very potent corticosteroid treatment was statistically significantly better than placebo for:

  • Investigator’s assessment (clear/nearly clear)at 2–4 weeks for clobetasol propionate once or twice daily [5 between-patient studies; 859 participants; very low quality evidence]73,125,167,216,227
  • Patient’s assessment (clear/nearly clear) at 2 weeks for clobetasol propionate twice daily [2 between-patient studies; 124 participants; low quality evidence]125,216

In people with psoriasis, there was no statistically significant difference between topical very potent corticosteroid treatment and placebo for:

  • Withdrawal due to adverse events at 2–4 weeks for clobetasol propionate once or twice daily [7 between-patient studies; 984 participants; very low quality evidence]32,73,125,167,174,216,227
  • Withdrawal due to lack of efficacy at 4 weeks for clobetasol propionate once or twice daily [3 studies (2 between- and 1 within-patient); 360 participants (385 randomised units); very low quality evidence]32,73,167
  • Skin atrophy at 4 weeks for clobetasol propionate once or twice daily [4 studies (3 between- and 1 within-patient); 439 participants (464 randomised units); very low quality evidence]32,73,167,174

8.1.5.3. Heterogeneity

  • For the outcome of investigator’s assessment of achieving clear/nearly clear status high heterogeneity was present between the results for the five studies. The heterogeneity could not be explained by any of the pre-specified subgroups for investigation or by excluding studies at high/very high risk of bias. It is likely to be caused by the small size of three of the studies167,216,227. The two sufficiently powered studies demonstrated a clear clinical benefit of very potent steroids compared with placebo.
  • There was no significant heterogeneity for the remaining outcomes.

8.1.6. Tazarotene vs. placebo

8.1.6.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsTazarotenePlaceboRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) – Tazarotene OD (follow-up 12 weeks)
2a
Weinstein 2003
randomised trialsvery seriousbseriouscno serious indirectnessseriousdnone50/860 (5.8%)9/443 (2%)RR 3.03 (0.83 to 11.07)41 more per 1000 (from 3 fewer to 205 more)⊕○○○
VERY LOW
Withdrawals due to adverse events – Tazarotene OD (follow-up 12 weeks)
3a
Weinstein 2003
Weinstein 1996
randomised trialsvery seriouseno serious inconsistencyno serious indirectnessno serious imprecisionnone112/1046 (10.7%)23/527 (4.4%)RR 2.45 (1.58 to 3.8)63 more per 1000 (from 25 more to 122 more)⊕⊕○○
LOW
Withdrawals due to lack of efficacy – Tazarotene OD (follow-up 12 weeks)
1
Weinstein 1996
randomised trialsseriousfno serious inconsistencyno serious indirectnessvery seriousgnone9/216 (4.2%)6/108 (5.6%)RR 0.75 (0.27 to 2.05)14 fewer per 1000 (from 41 fewer to 58 more)⊕○○○
VERY LOW
Skin atrophy – Tazarotene OD (follow-up 12 weeks)
1
Weinstein 1996
randomised trialsseriousfno serious inconsistencyno serious indirectnessno serious imprecisionnone0/216 (0%)0/108 (0%)not poolednot pooled⊕⊕⊕○
MODERATE
a

Two studies reported within one publication

b

2/2 unclear allocation concealment and blinding; 2/2 high drop-out rate (tazarotene: 38.5% and 36.6%; placebo: 32.2% and 23.8%)

c

Heterogeneity was present (I2 = 61%) that could not be explained by pre-defined subgroups (however, both studies showed the same direction of effect)

d

Confidence interval ranges from clinically important effect to no effect

e

3/3 unclear allocation concealment; 2/3 (weighted 47.4 and 39.1%) unclear blinding and high drop-out rate (tazarotene: 38.5% and 36.6%; placebo: 32.2% and 23.8%)

f

Unclear allocation concealment

g

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

8.1.6.2. Evidence statements

In people with psoriasis, placebo was statistically significantly better than tazarotene applied once daily for:

  • Withdrawal due to adverse events at 12 weeks [3 between-patient studies; 1573 participants; low quality evidence] 418420

In people with psoriasis, there were no events with either tazarotene or placebo for:

  • Skin atrophy at 12 weeks [1 between-patient study; 324 participants; moderate quality evidence]418,420

In people with psoriasis, there was no statistically significant difference between tazarotene and placebo applied once daily for:

  • Investigator’s assessment (clear/nearly clear) at 12 weeks [2 between-patient studies; 1303 participants; very low quality evidence]419
  • Withdrawal due to lack of efficacy at 12 weeks [1 between-patient study; 324 participants; very low quality evidence]418,420

8.1.6.3. Subgroups and heterogeneity

  • For the outcome of investigator’s assessment of achieving clear/nearly clear status heterogeneity was present between the results. The heterogeneity could not be explained by any of the pre-specified subgroups for investigation or excluding studies at high risk of bias.
  • There was no significant heterogeneity for the remaining outcomes.

8.1.7. Potent corticosteroid vs. placebo for maintenance of remission

This study included participants who achieved remission after 3–4 weeks treatment with betamethasone dipropionate (remission defined as: erythema score ≤ 1 (slight or minimal); induration = 0.5 (none-slight); scaling = 0 (none)). The maintenance regimen for those in remission and randomised to active treatment was intermittent betamethasone dipropionate applied to the site of the healed lesion (three consecutive applications 12 hours apart, once a week for a maximum treatment period of 6 months).

8.1.7.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCorticosteroid (potent)PlaceboRelative (95% CI)Absolute
Investigator’s assessment (maintaining clear/slight) – intermittent betamethasone dipropionate BD (follow-up 24 weeks)
1
Katz 1991
randomised trialsseriousano serious inconsistencyseriousbno serious imprecisionnone27/46 (58.7%)7/44 (15.9%)RR 3.69 (1.79 to 7.59)428 more per 1000 (from 126 more to 1000 more)⊕⊕○○
LOW
Time-to-relapse – intermittent betamethasone dipropionate BD (follow-up 24 weeks)
1
Katz 1991
randomised trialsseriousano serious inconsistencyseriouscno serious imprecisionnone16/46 (34.8%)35/44 (79.5%)HR 0.37 (0.21 to 0.67)351 fewer per 1000 (from 141 fewer to 512 fewer)⊕⊕○○
LOW
Withdrawals due to adverse events – intermittent betamethasone dipropionate BD (follow-up 24 weeks)
1
Katz 1991
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone0/44 (0%)0/42 (0%)not poolednot pooled⊕⊕⊕○
MODERATE
Skin atrophy – intermittent betamethasone dipropionate BD (follow-up 24 weeks)
1
Katz 1991
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone0/46 (0%)0/44 (0%)not poolednot pooled⊕⊕⊕○
MODERATE
a

Unclear allocation concealment and blinding

b

Definition of response does not match the review criteria for clear/nearly clear (broader - clear or slight on a 4-point scale; clear, slight, moderate, severe) and so may overestimate efficacy

c

Definition of relapse includes failure just at target plaques or in overall disease status

8.1.7.2. Evidence statements

In people with psoriasis, intermittent twice daily topical potent corticosteroid (betamethasone dipropionate) was statistically significantly better than placebo for the maintenance of remission for:

  • Investigator’s assessment (clear/slight) at 24 weeks [1 between-patient study; 90 participants; low quality evidence]178
  • Time-to-relapse after a maximum follow-up of at 24 weeks [1 between-patient study; 90 participants; low quality evidence]178

In people with psoriasis, there were no events with either intermittent twice daily topical potent corticosteroid (betamethasone dipropionate) or placebo for the maintenance of remission for:

  • Withdrawal due to adverse events at 24 weeks [1 between-patient study; 86 participants; moderate quality evidence]178
  • Skin atrophy at 24 weeks [1 between-patient study; 90 participants; moderate quality evidence]178

8.1.7.3. Heterogeneity

Not applicable as only one study assessed potent corticosteroid compared with placebo for the maintenance of remission.

8.1.8. Vitamin D or vitamin D analogue vs. potent corticosteroid

8.1.8.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D or vitamin D analoguesCorticosteroid (potent)Relative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) – Calcipotriol OD/BD or calcitriol BD vs betamethasone dipropionate OD/BD or betamethasone valerate BD (follow-up 4–8 weeks)
6
Fleming 2010A
Kaufmann 2002
Douglas 2002
Papp 2003
Molin 1997
Camarasa 2003
randomised trialsseriousavery seriousbno serious indirectnessseriouscnone547/1565 (35%)730/1571 (46.5%)RR 0.76 (0.62 to 0.94)122 fewer per 1000 (from 28 fewer to 177 fewer)⊕○○○
VERY LOW
Patient’s assessment (clear/nearly clear) - Calcipotriol OD vs betamethasone dipropionate OD (follow-up 4 weeks)
1
Kaufmann 2002
randomised trialsseriousdno serious inconsistencyno serious indirectnessno serious imprecisionnone137/480 (28.5%)216/476 (45.4%)RR 0.63 (0.53 to 0.75)168 fewer per 1000 (from 113 fewer to 213 fewer)⊕⊕⊕○
MODERATE
Patient’s assessment (clear/nearly clear) - Calcipotriol BD vs betamethasone dipropionate BD (follow-up 4 weeks)
1
Douglas 2002
randomised trialsseriousdno serious inconsistencyno serious indirectnessseriouscnone140/365 (38.4%)183/363 (50.4%)RR 0.76 (0.64 to 0.9)121 fewer per 1000 (from 50 fewer to 181 fewer)⊕⊕○○
LOW
Patient’s assessment (clear/nearly clear) - Calcipotriol BD vs betamethasone valerate BD (follow-up 6 weeks)
2
Cunliffe 1992
Kragballe 1991
randomised trialsseriousdno serious inconsistencyno serious indirectnessseriouscnone403/543 (61.2%)338/542 (50.5%)RR 1.19 (1.10 to 1.29)118 more per 1000 (from 62 more to 181 more)⊕⊕○○
LOW
% change in PASI - Calcipotriol (BD) vs betamethasone valerate (BD) (follow-up 6–8 weeks; Better indicated by lower values)
2
Kragballe 1991
Molin 1997
randomised trialsseriouseno serious inconsistencyno serious indirectnessno serious imprecisionnone547549-MD 5.94 higher (2.29 to 9.60 higher)⊕⊕⊕○
MODERATE
Relapse rate (requiring re-treatment [not maintaining clear/nearly clear] within 8-weeks post Tx) - Calcitriol BD vs betamethasone dipropionate BD
1
Camarasa 2003
randomised trialsvery seriousfno serious inconsistencyseriousgserioushnone30/58 (51.7%)55/73 (75.3%)RR 0.69 (0.52 to 0.91)234 fewer per 1000 (from 68 fewer to 362 fewer)⊕○○○
VERY LOW
Mean time to relapse (requiring re-treatment [not maintaining clear/nearly clear] within 8-weeks post Tx) - Calcitriol BD vs betamethasone dipropionate BD
1
Camarasa 2003
randomised trialsvery seriousfno serious inconsistencyno serious indirectnessseriousinone5873-Vitamin D: 25.3 days

Corticosteroid: 23.4 days
⊕○○○
VERY LOW
Withdrawals due to adverse events – Calcipotriol OD/BD or calcitriol BD vs betamethasone dipropionate OD/BD, betamethasone valerate BD or fluocinonide BD (follow-up 4–8 weeks)
7
Douglas 2002
Kaufmann 2002
Cunliffe 1992
Kragballe 1991
Molin 1997
Bruce 1994
Camarasa 2003
randomised trialsseriousjno serious inconsistencykno serious indirectnessseriouscnone30/1709 (1.8%)14/1718 (0.81%)RR 2.10 (1.13 to 3.90)9 more per 1000 (from 1 more to 24 more)⊕⊕○○
LOW
Withdrawals due to lack of efficacy – Calcipotriol or calcitriol BD vs betamethasone dipropionate or valerate BD (follow-up 6 weeks)
3
Cunliffe 1992
Kragballe 1991
Camarasa 2003
randomised trialsseriouslno serious inconsistencymno serious indirectnessvery seriousnnone11/661 (1.7%)11/660 (1.7%)RR 1 (0.44 to 2.28)0 fewer per 1000 (from 9 fewer to 21 more)⊕○○○
VERY LOW
Skin atrophy – Calcipotriol BD vs betamethasone dipropionate or valerate BD (follow-up 4–8 weeks)
2
Papp 2003
Molin 1997
randomised trialsseriousono serious inconsistencyno serious indirectnessvery seriousnnone0/515 (0%)5/523 (0.96%)RR 0.17 (0.02 to 1.4)8 fewer per 1000 (from 9 fewer to 4 more)⊕○○○
VERY LOW
a

6/6 unclear allocation concealment; 2/6 (26.8% weighted) unclear blinding

b

Heterogeneity was present (I2 = 81%) that could not be explained by pre-defined subgroups (however, 5/6 studies showed the same direction of effect)

c

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit in favour of corticosteroid to no clinically important difference)

d

Unclear allocation concealment

e

2/2 unclear allocation concealment; 1/2 (26.2% weighted) unclear blinding and unclear baseline demographics

f

Unclear allocation concealment and blinding; also, unclear baseline comparability as only includes those in each group who achieved remission; therefore, there are fewer participants in the vitamin D or vitamin D analogue group

g

Surrogate outcome for duration of remission and definition of relapse = requiring re-treatment (not maintaining clear/nearly clear)

h

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit in favour of vitamin D or vitamin D analogue to no clinically important difference)

i

No SD given

j

7/7 unclear allocation concealment; 4/7 unclear blinding (55.5% weighted); 1/7 (22% weighted) unclear baseline demographics; 1/7 (11.2% weighted) dropout rate not stratified by group

k

No statistically significant heterogeneity but one study (Bruce) favours a different treatment

l

3/3 unclear allocation concealment; 2/3 (81.8% weighted) unclear blinding

m

No statistically significant heterogeneity but one study (Kragballe) favours a different treatment

n

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

o

2/2 unclear allocation concealment; 1/2 (58.4% weighted) unclear blinding and unclear baseline demographics

8.1.8.2. Evidence statements

In people with psoriasis, potent corticosteroid was statistically significantly better than vitamin D or vitamin D analogue for:

  • Investigator’s assessment (clear/nearly clear) at 4–8 weeks for calcipotriol once or twice daily or calcitriol twice daily compared to betamethasone dipropionate once or twice daily or betamethasone valerate twice daily [6 between-patient studies; 3136 participants; very low quality evidence]49,79,102,179,255,302
  • Patient’s assessment (clear/nearly clear) at 4 weeks for calcipotriol once or twice daily compared to betamethasone dipropionate once or twice daily [2 between-patient studies; 1684 participants; low to moderate quality evidence]79,179
  • Withdrawals due to adverse events at 4–8 weeks for calcipotriol once or twice daily or calcitriol twice daily compared to betamethasone dipropionate once or twice daily, betamethasone valerate twice daily or fluocinonide twice daily [7 studies (6 between- and 1 within-patient); 3082 participants (3427 randomised units); low quality evidence]47,49,66,79,179,198,255

In people with psoriasis, vitamin D or vitamin D analogue was statistically significantly better than potent corticosteroid for:

  • Patient’s assessment (clear/nearly clear) at 6 weeks for calcipotriol twice daily compared to betamethasone valerate twice daily [2 studies (1 between- and 1 within-patient); 743 participants (1085 randomised units); low quality evidence]66,198
  • % change in PASI at 6–8 weeks for calcipotriol twice daily compared to betamethasone valerate twice daily [2 studies (1 between- and 1 within-patient); 754 participants (1096 randomised units); moderate quality evidence]198,255
  • Relapse rate (requiring re-treatment [not maintaining clear/nearly clear] within 8-weeks post treatment) for calcitriol twice daily compared with betamethasone dipropionate twice daily [1 between-patient study; 131 participants; very low quality evidence]49

In people with psoriasis, there was no statistically significant difference between potent corticosteroid and vitamin D or vitamin D analogue for:

  • Withdrawals due to lack of efficacy at 6 weeks for calcipotriol or calcitriol twice daily compared with betamethasone dipropionate or valerate twice daily [3 studies (1 between- and 2 within-patient); 976 participants (1321 randomised units); very low quality evidence] 49,66,198
  • Skin atrophy at 4–8 weeks for calcipotriol twice daily vs betamethasone dipropionate or valerate twice daily [2 between-patient studies; 1038 participants; very low quality evidence]255,302
Evidence statement for individual study where no statistical analysis could be performed

In people with psoriasis, vitamin D or vitamin D analogue was better than potent corticosteroid for:

  • Mean time to relapse (requiring re-treatment [not maintaining clear/nearly clear] within 8-weeks post treatment) for calcitriol twice daily compared with betamethasone dipropionate twice daily [1 between-patient study; 131 participants; very low quality evidence]49

8.1.8.3. Heterogeneity

  • For the outcome of investigator’s assessment of achieving clear/nearly clear status heterogeneity was present. The heterogeneity could not be explained by any of the pre-specified subgroups for investigation or by excluding studies at higher risk of bias.
  • For the outcome of patient’s assessment of achieving clear/nearly clear status heterogeneity was present. The heterogeneity was explained by creating subgroups based on treatment frequency and the specific agent, suggesting that betamethasone valerate may be less effective than betamethasone dipropionate.
  • There was no significant heterogeneity for the remaining outcomes.

8.1.9. Concurrent vitamin D or vitamin D analogue and potent corticosteroid (one in the morning and one in the evening) vs. vitamin D or vitamin D analogue alone

8.1.9.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsConcurrent vitamin D or analogues and potent corticosteroidVitamin D or vitamin D analogueRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) - Calcipotriol and betamethasone valerate vs calcipotriol OD (follow-up 8 weeks)
1
Kragballe 1998
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionNone94/174 (54%)49/172 (28.5%)RR 1.9 (1.44 to 2.49)256 more per 1000 (from 125 more to 424 more)⊕⊕⊕○
MODERATE
Investigator’s assessment (clear/nearly clear) - Calcipotriol and betamethasone valerate vs calcipotriol BD (follow-up 6–8 weeks)
2
Kragballe 1998
Ruzicka 1998
randomised trialsseriousbno serious inconsistencyno serious indirectnessseriouscNone154/252 (61.1%)121/258 (46.9%)RR 1.32 (1.12 to 1.54)150 more per 1000 (from 56 more to 253 more)⊕⊕○○
LOW
Investigator’s assessment (clear/nearly clear among those who did not respond to calcipotriol after 2 weeks) - Calcipotriol and betamethasone valerate vs calcipotriol BD (follow-up 6 weeks)
1
Ruzicka 1998
randomised trialsseriousdno serious inconsistencyno serious indirectnessseriouscNone27/39 (69.2%)22/49 (44.9%)RR 1.54 (1.06 to 2.24)242 more per 1000 (from 27 more to 557 more)⊕⊕○○
LOW
Patient’s assessment (clear/nearly clear) - Calcipotriol and betamethasone valerate vs calcipotriol OD (follow-up 8 weeks)
1
Kragballe 1998
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionNone89/174 (51.1%)46/172 (26.7%)RR 1.91 (1.44 to 2.55)243 more per 1000 (from 118 more to 415 more)⊕⊕⊕○
MODERATE
Patient’s assessment (clear/nearly clear) - Calcipotriol and betamethasone valerate vs calcipotriol BD (follow-up 8 weeks)
1
Kragballe 1998
randomised trialsseriousano serious inconsistencyno serious indirectnessseriouscNone89/174 (51.1%)69/172 (40.1%)RR 1.28 (1.01 to 1.61)112 more per 1000 (from 4 more to 245 more)⊕⊕○○
LOW
Withdrawals due to adverse events - Calcipotriol and betamethasone valerate vs calcipotriol OD (follow-up 8 weeks)
1
Kragballe 1998
randomised trialsseriousano serious inconsistencyno serious indirectnessvery seriouseNone3/168 (1.8%)8/163 (4.9%)RR 0.36 (0.1 to 1.35)31 fewer per 1000 (from 44 fewer to 17 more)⊕○○○
VERY LOW
Withdrawals due to adverse events - Calcipotriol and corticosteroid (betamethasone valerate or diflucortolone valerate) vs calcipotriol BD (follow-up 4–8 weeks)
3
Kragballe 1998
Ruzicka 1998
Salmhofer 2000
randomised trialsfseriousgno serious inconsistencyno serious indirectnessvery seriouseNone4/308 (1.3%)8/303 (2.6%)RR 0.52 (0.17 to 1.61)13 fewer per 1000 (from 22 fewer to 16 more)⊕○○○
VERY LOW
Withdrawals due to lack of efficacy - Calcipotriol and betamethasone valerate vs calcipotriol OD (follow-up 8 weeks)
1
Kragballe 1998
randomised trialsseriousano serious inconsistencyno serious indirectnessvery seriouseNone1/166 (0.6%)2/174 (1.1%)RR 0.52 (0.05 to 5.73)6 fewer per 1000 (from 11 fewer to 54 more)⊕○○○
VERY LOW
Withdrawals due to lack of efficacy - Calcipotriol and betamethasone/diflucortolone valerate vs calcipotriol BD (follow-up 4–8 weeks)
2
Kragballe 1998
Salmhofer 2000
randomised trialsfseriousano serious inconsistencyno serious indirectnessvery seriouseNone1/229 (0.44%)3/223 (1.3%)RR 0.32 (0.03 to 3.06)9 fewer per 1000 (from 13 fewer to 28 more)⊕○○○
VERY LOW
a

Unclear allocation concealment and blinding

b

2/2 unclear allocation concealment and blinding; 1/2 includes only patients with at least 4 weeks therapy, but this means just 2 weeks randomised

c

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit of concurrent treatment to no clinically important difference)

d

Unclear allocation concealment and blinding; includes only patients with at least 4 weeks therapy, but this means just 2 weeks randomised

e

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

f

Data for Salmhofer are from a published Cochrane Review

g

3/3 unclear allocation concealment and blinding; 1/3 includes only patients with at least 4 weeks therapy, but this means just 2 weeks randomised

8.1.9.2. Evidence statements

In people with psoriasis, concurrent vitamin D or vitamin D analogue and potent corticosteroid treatment (one applied in the morning and one in the evening) was statistically significantly better than vitamin D or vitamin D analogue alone for:

  • Investigator’s assessment (clear/nearly clear)at 6–8 weeks for calcipotriol and betamethasone valerate compared with calcipotriol once or twice daily [2 between-patient studies; 682 participants; low to moderate quality evidence]197,344
  • Investigator’s assessment (clear/nearly clear among those who did not respond to calcipotriol after 2 weeks) at 6 weeks for calcipotriol and betamethasone valerate compared with calcipotriol twice daily [1 between-patient study; 88 participants; low quality evidence]344
  • Patient’s assessment (clear/nearly clear) at 8 weeks for calcipotriol and betamethasone valerate compared with calcipotriol once or twice daily [1 between-patient study; 518 participants; low to moderate quality evidence]197

In people with psoriasis, there was no statistically significant difference between concurrent vitamin D or vitamin D analogue and potent corticosteroid treatment (one applied in the morning and one in the evening) and vitamin D or vitamin D analogue alone for:

  • Withdrawals due to adverse events at 4–8 weeks for calcipotriol and betamethasone valerate or diflucortolone valerate compared with calcipotriol once or twice daily [3 studies (2 between- and 1 within-patient); 711 participants (774 randomised units); very low quality evidence]197,344,346
  • Withdrawals due to lack of efficacy calcipotriol and betamethasone valerate or diflucortolone valerate compared with calcipotriol once or twice daily [2 studies (1 between- and 1 within-patient); 563 participants (626 randomised units); very low quality evidence]197,346

8.1.9.3. Heterogeneity

  • For the outcomes of investigator’s and patient’s assessment of achieving clear/nearly clear status heterogeneity was present. The heterogeneity was removed by separating into subgroups based on frequency of administration of vitamin D or vitamin D analogue, suggesting that concurrent use of vitamin D or vitamin D analogue and potent steroid (one applied in the morning and one in the evening) is clinically more effective than once daily vitamin D or vitamin D analogue alone, but the effect in favour of the concurrent use is smaller compared with twice daily vitamin D or vitamin D analogue application.
  • There was no significant heterogeneity for the remaining outcomes but OD and BD subgroups were kept separate where necessary to avoid double counting data from the Kragballe1998 study.

8.1.10. Combined product containing vitamin D or vitamin D analogue and potent corticosteroid (calcipotriol plus betamethasone dipropionate) vs. vitamin D or vitamin D analogue alone

8.1.10.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCombined productVitamin D or vitamin D analogueRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) – Combination OD vs. vitamin D or vitamin D analogue (calcipotriol or tacalcitol) OD (follow-up 4–8 weeks)
4
Fleming 2010A
Kaufmann 2002
Langley 2011 A
Ortonne 2004
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionNone536/1084 (49.4%)192/995 (19.3%)RR 2.65 (2.3 to 3.05)318 more per 1000 (from 251 more to 396 more)⊕⊕⊕○
MODERATE
Investigator’s assessment (clear/nearly clear) - Combination OD vs. vitamin D or vitamin D analogue (calcipotriol) BD (follow-up 4–8 weeks)
2
Guenther 2002
Kragballe 2004
randomised trialsseriousbno serious inconsistencyno serious indirectnessno serious imprecisionNone273/472 (57.8%)248/554 (44.8%)RR 1.31 (1.16 to 1.48)139 more per 1000 (from 72 more to 215 more)⊕⊕⊕○
MODERATE
Patient’s assessment (clear/nearly clear) - Combination OD vs. vitamin D or vitamin D analogue (calcipotriol or tacalcitol) OD or BD (follow-up 4–8 weeks)
4
Kaufmann 2002
Guenther 2002
Langley 2011 A
Ortonne 2004
randomised trialsseriouscvery seriousdno serious indirectnessno serious imprecisionNone628/1060 (59.2%)333/1122 (29.7%)RR 2.05 (1.35 to 3.11)312 more per 1000 (from 104 more to 626 more)⊕○○○
VERY LOW
% change in PASI – Combination OD vs. vitamin D or vitamin D analogue (calcipotriol or tacalcitol) OD or BD (follow-up 4–8 weeks; Better indicated by lower values)
5
Fleming 2010A
Kaufmann 2002
Kragballe 2004
Guenther 2002
Langley 2011 A
randomised trialsseriouseno serious inconsistencyno serious indirectnessno serious imprecisionNone10371297-MD 11.62 lower (14.87 to 8.37 lower)⊕⊕⊕○
MODERATE
Relapse rate at 8 weeks post-treatment - Combination OD vs. tacalcitol OD (follow-up 8 weeks + 8 weeks post-treatment)
1
Langley 2011 A
randomised trialsvery seriousfno serious inconsistencyseriousgserioushNone28/67 (41.8%)7/31 (22.6%)RR 1.85 (0.91 to 3.77)192 more per 1000 (from 20 fewer to 625 more)⊕○○○
VERY LOW
Median time to relapse – Combination OD vs. tacalcitol OD (follow-up 8 weeks + 8 weeks post-treatment)
1
Langley 2011 A
randomised trialsvery seriousfno serious inconsistencyno serious indirectnessseriousiNone6731-Combination: 63 days Vitamin D: 61 days⊕○○○
VERY LOW
Withdrawals due to adverse events – Combination OD vs. vitamin D or vitamin D analogue (calcipotriol or tacalcitol) OD or BD (follow-up 4–8 weeks)
3
Kaufmann 2002
Guenther 2002
Langley 2011 A
randomised trialsseriousjno serious inconsistencyno serious indirectnessno serious imprecisionNone6/797 (0.75%)23/839 (2.7%)RR 0.28 (0.12 to 0.67)20 fewer per 1000 (from 9 fewer to 24 fewer)⊕⊕⊕○
MODERATE
Withdrawals due to lack of efficacy - Combination OD vs. calcipotriol BD (follow-up 4 weeks)
1
Guenther 2002
randomised trialsseriouskno serious inconsistencyno serious indirectnessvery seriouslNone0/151 (0%)2/227 (0.9%)RR 0.3 (0.01 to 6.21)6 fewer per 1000 (from 9 fewer to 46 more)⊕○○○
VERY LOW
Skin atrophy - Combination OD vs. calcipotriol BD (follow-up 4–12 weeks)
2
Kragballe 2004
Guenther 2002
randomised trialsseriousmno serious inconsistencyseriousnvery seriouslNone2/473 (0.42%)1/554 (0.18%)RR 2.09 (0.27 to 16.53)2 more per 1000 (from 1 fewer to 28 more)⊕○○○
VERY LOW
a

4/4 unclear allocation concealment; 1/4 single blind; 4/4 differential dropout (higher with vitamin D or vitamin D analogue, but acceptable level in all but 1 study)

b

2/2 unclear allocation concealment; 1/2 (59.1% weighted) double blind in combination arm but single blind (investigator) in vitamin D or vitamin D analogue group

c

4/4 unclear allocation concealment; 1/4 single blind (investigator); 3/4 differential dropout rate (but only >20% in one study)

d

Heterogeneity was present (I2 = 93%) that could not be explained by pre-defined subgroups (however, all studies showed the same direction of effect)

e

5/5 unclear allocation concealment; 1/5 (13.8% weighted) single blind (investigator); 1/5 (35.2% weighted) double blind in combination arm but single blind (investigator) in vitamin D or vitamin D analogue group; 3/5 differential dropout (but none >20%)

f

Unclear allocation concealment and differential dropout rate (higher in vitamin D or vitamin D analogue group but not >20%); also, unclear baseline comparability as only includes those in each group who achieved remission; therefore, there are fewer participants in the vitamin D or vitamin D analogue alone group

g

Surrogate outcome for duration of remission

h

Confidence interval ranges from clinically significant effect to no effect

i

No range given

j

3/3 unclear allocation concealment; 1/3 (17.7% weighted) single blind (investigator); 2/3 differential dropout rate (but not >20%)

k

Unclear allocation concealment

l

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

m

2/2 unclear allocation concealment; 1/2 (38.3% weighted) double blind in combination arm but single blind (investigator) in vitamin D or vitamin D analogue group and differential dropout (but not >20%)

n

Data are for full study period (so combination group received vitamin D or vitamin D analogue only for the final 4 of 12 weeks)

8.1.10.2. Evidence statements

In people with psoriasis, a combined product containing calcipotriol monohydrate and betamethasone dipropionate once daily was statistically significantly better than calcipotriol once or twice daily or tacalcitol once daily for:

  • Investigator’s assessment (clear/nearly clear)at 4–8 weeks [6 between-patient studies; 1249 participants; moderate quality evidence]102,132,179,201,208,298
  • Patient’s assessment (clear/nearly clear) at 4–8 weeks [4 between-patient studies; 2182 participants; very low quality evidence]132,179,208,298
  • Percentage change in PASI at 4–8 weeks [5 between-patient studies; 2334 participants; moderate quality evidence]102,132,179,201,208
  • Withdrawals due to adverse events at 4–8 weeks [3 between-patient studies; 1636 participants; moderate quality evidence]132,179,208

In people with psoriasis, there was no statistically significant difference between a combined product containing calcipotriol monohydrate and betamethasone dipropionate once daily and vitamin D or vitamin D analogue once or twice daily for:

  • Relapse rate at 8 weeks post-treatment for the combination product compared with tacalcitol once daily [1 between-patient study; 98 participants; very low quality evidence]208
  • Withdrawals due to lack of efficacy at 4 weeks for the combination product compared with calcipotriol twice daily [1 between-patient study; 378 participants; very low quality evidence]132
  • Skin atrophy at 4–12 weeks for the combination product compared with calcipotriol twice daily [2 between-patient studies; 1027 participants; very low quality evidence]132,201
Evidence statement for individual study where no statistical analysis could be performed

In people with psoriasis, a combined product containing calcipotriol monohydrate and betamethasone dipropionate once daily was better than vitamin D or vitamin D once daily for:

  • Median time to relapse at 8 weeks post-treatment among those who had achieved remission with the combination product compared with tacalcitol once daily [1 between-patient study; 98 participants; very low quality evidence]208

8.1.10.3. Heterogeneity

  • For the outcome of investigator’s assessment of achieving clear/nearly clear status heterogeneity was present. The heterogeneity was removed by separating into subgroups based on frequency of administration of vitamin D or vitamin D analogue, suggesting that use of combined vitamin D or vitamin D analogue and potent steroid is clinically more effective than once daily vitamin D or vitamin D analogue alone, but the effect in favour of the combined use was smaller compared with twice daily vitamin D or vitamin D analogue application.
  • For the outcome of patient’s assessment of achieving clear/nearly clear status high heterogeneity was present. The heterogeneity was not fully explained by any of the pre-specified subgroups although for the comparison with once daily vitamin D or vitamin D analogue the combination was clearly clinically more effective in all studies, but again the effect in favour of the combined use was smaller compared with twice daily vitamin D or vitamin D analogue application.
  • There was no significant heterogeneity for the remaining outcomes.

8.1.11. Combined product containing vitamin D or vitamin D analogue and potent corticosteroid (calcipotriol plus betamethasone dipropionate) vs. potent corticosteroid

8.1.11.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D and corticosteroid combinationPotent corticosteroidRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) – combination OD vs betamethasone dipropionate OD (follow-up 4–8 weeks)
2
Fleming 2010A
Kaufmann 2002
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionNone320/652 (49.1%)190/559 (34%)RR 1.53 (1.33 to 1.76)180 more per 1000 (from 112 more to 258 more)⊕⊕⊕○
MODERATE
Patient’s assessment (clear/nearly clear) – combination OD vs betamethasone dipropionate OD (follow-up 4 weeks)
1
Kaufmann 2002
randomised trialsseriousbno serious inconsistencyno serious indirectnessno serious imprecisionNone316/490 (64.5%)216/476 (45.4%)RR 1.42 (1.26 to 1.6)191 more per 1000 (from 118 more to 272 more)⊕⊕⊕○
MODERATE
% change in PASI – combination OD vs betamethasone dipropionate OD (follow-up 4–8 weeks; Better indicated by lower values)
2
Fleming 2010A
Kaufmann 2002
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionNone652559-MD 9.94 lower (15.75 to 4.14 lower)⊕⊕⊕○
MODERATE
Withdrawals due to adverse events – combination OD vs betamethasone dipropionate OD (follow-up 4 weeks)
1
Kaufmann 2002
randomised trialsseriousbno serious inconsistencyno serious indirectnessvery seriouscNone3/480 (0.63%)5/452 (1.1%)RR 0.56 (0.14 to 2.35)5 fewer per 1000 (from 10 fewer to 15 more)⊕○○○
VERY LOW
a

2/2 unclear allocation concealment

b

Unclear allocation concealment

c

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

8.1.11.2. Evidence statements

In people with psoriasis, a combined product containing calcipotriol monohydrate and betamethasone dipropionate was statistically significantly better than potent corticosteroid (betamethasone dipropionate once daily) for:

  • Investigator’s assessment (clear/nearly clear) at 4–8 weeks [2 between-patient studies; 1211 participants; moderate quality evidence]102,179
  • Patient’s assessment (clear/nearly clear) at 4 weeks [1 between-patient study; 966 participants; moderate quality evidence]179
  • Percentage change in PASI at 4–8 weeks [2 between-patient studies; 1211 participants; moderate quality evidence] 102,179

In people with psoriasis, there was no statistically significant difference between a combined product containing calcipotriol monohydrate and betamethasone dipropionate and potent corticosteroid (betamethasone dipropionate once daily) for:

  • Withdrawals due to adverse events at 4 weeks [1 between-patient study; 932 participants; very low quality evidence]179

8.1.11.3. Heterogeneity

  • There was no significant heterogeneity for the any of the outcomes.

8.1.12. Combined product containing vitamin D or vitamin D analogue and potent corticosteroid (calcipotriol plus betamethasone dipropionate) then vitamin D or vitamin D analogue vs. vitamin D or vitamin D analogue alone

8.1.12.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D and corticosteroid combination then vitamin DVitamin DRelative (95% CI)Absolute
Investigator's assessment (clear/nearly clear) – Combination (OD) (8 wk) then calcipotriol OD (4 wk) vs. calcipotriol BD (12 wk) (follow-up 12 weeks)
1 Kragballe 2004randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone178/322 (55.3%)133/327 (40.7%)RR 1.36 (1.15 to 1.6)146 more per 1000 (from 61 more to 244 more)⊕⊕⊕○
MODERATE
Investigator's assessment (clear/nearly clear) -Combination (OD) (4 wk) then calcipotriol OD weekdays/combination weekends (8 wks) vs. calcipotriol BD (12 wk) (follow-up 12 weeks)
1 Kragballe 2004randomised trialsseriousano serious inconsistencyno serious indirectnessseriousbnone154/323 (47.7%)133/327 (40.7%)RR 1.17 (0.99 to 1.39)69 more per 1000 (from 4 fewer to 159 more)⊕⊕○○
LOW
Investigator's assessment (clear/nearly clear) -Combination (OD) (4 wk) then calcipotriol OD (4 wks) vs. tacalcitol OD (8 wk) (follow-up 8 weeks)
1 Ortonne 2004randomised trialsseriouscno serious inconsistencyno serious indirectnessno serious imprecisionnone126/249 (50.6%)59/252 (23.4%)RR 2.16 (1.68 to 2.79)272 more per 1000 (from 159 more to 419 more)⊕⊕⊕○
MODERATE
Patient's assessment (clear/nearly clear) -Combination (OD) (4 wk) then calcipotriol OD (4 wks) vs. tacalcitol OD (8 wk) (follow-up 8 weeks)
1
Ortonne 2004
randomised trialsseriouscno serious inconsistencyno serious indirectnessno serious imprecisionnone130/249 (52.2%)68/252 (27%)RR 1.93 (1.53 to 2.45)251 more per 1000 (from 143 more to 391 more)⊕⊕⊕○
MODERATE
% change in PASI -Combination (OD) (8 wk) then calcipotriol OD (4 wk) vs. calcipotriol BD (12 wk) (follow-up 12 weeks; Better indicated by lower values)
1
Kragballe 2004
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone322327-MD 9.2 lower (14.68 to 3.72 lower)⊕⊕⊕○
MODERATE
% change in PASI -Combination (OD) (4 wk) then vitamin D or vitamin D analogue OD weekdays/combination OD weekends (8 wks) vs. calcipotriol BD (12 wk) (follow-up 12 weeks; Better indicated by lower values)
1
Kragballe 2004
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone323327-MD 4.4 lower (8.35 to 0.45 lower)⊕⊕⊕○
MODERATE
% change in PASI -Combination (OD) (4 wk) then calcipotriol OD (4 wks) vs. tacalcitol OD (8 wk) (follow-up 8 weeks; Better indicated by lower values)
1
Ortonne 2004
randomised trialsseriouscno serious inconsistencyno serious indirectnessno serious imprecisionnone249252-MD 20.6 lower (32.87 to 8.33 lower)⊕⊕⊕○
MODERATE
Withdrawal due to adverse events -Combination (OD) (4 wk) then calcipotriol BD (8 wk) vs calcipotriol BD (12 wk) (follow-up 12 weeks)
1
Saraceno 2007
randomised trialsvery seriousdno serious inconsistencyno serious indirectnessvery seriousenone3/53 (5.7%)2/48 (4.2%)RR 1.36 (0.24 to 7.79)15 more per 1000 (from 32 fewer to 283 more)⊕○○○
VERY LOW
Withdrawal due to adverse events -Combination (OD) (4 wk) then calcipotriol OD (4 wks) vs. tacalcitol OD (8 wk) (follow-up 8 weeks)
1
Ortonne 2004
randomised trialsseriouscno serious inconsistencyno serious indirectnessvery seriousenone6/223 (2.7%)11/228 (4.8%)RR 0.56 (0.21 to 1.48)21 fewer per 1000 (from 38 fewer to 23 more)⊕○○○
VERY LOW
Withdrawal due to lack of efficacy -Combination (OD) (4 wk) then calcipotriol BD (8 wk) vs calcipotriol BD (12 wk) (follow-up 12 weeks)
1
Saraceno 2007
randomised trialsvery seriousdno serious inconsistencyno serious indirectnessvery seriousenone1/51 (2%)3/49 (6.1%)RR 0.32 (0.03 to 2.98)42 fewer per 1000 (from 59 fewer to 121 more)⊕○○○
VERY LOW
Withdrawal due to lack of efficacy -Combination (OD) (4 wk) then calcipotriol OD (4 wks) vs. tacalcitol OD (8 wk) (follow-up 8 weeks)
1
Ortonne 2004
randomised trialsseriouscno serious inconsistencyno serious indirectnessvery seriousenone3/220 (1.4%)8/225 (3.6%)RR 0.38 (0.1 to 1.43)22 fewer per 1000 (from 32 fewer to 15 more)⊕○○○
VERY LOW
Skin atrophy -Combination (OD) (8 wk) then calcipotriol OD (4 wk) vs. calcipotriol BD (12 wk) (follow-up 12 weeks)
1
Kragballe 2004
randomised trialsseriousano serious inconsistencyno serious indirectnessvery seriousenone1/322 (0.31%)0/327 (0%)RR 3.05 (0.12 to 74.51)-⊕○○○
VERY LOW
Skin atrophy -Combination (OD) (4 wk) then calcipotriol OD weekdays/combination OD weekends (8 wks) vs. calcipotriol BD (12 wk) (follow-up 12 weeks)
1
Kragballe 2004
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone0/322 (0%)0/327 (0%)not poolednot pooled⊕⊕⊕○
MODERATE
a

Unclear allocation concealment and calcipotriol group only single blind (investigator)

b

Confidence interval ranges from clinically important effect to no effect

c

Unclear allocation concealment and high differential dropout (15.7% in combination group and 20.2% in tacalcitol group)

d

Unblinded and high dropout rate (33.3% in combination group and 38.7% in calcipotriol group)

e

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

8.1.12.2. Evidence statements

In people with psoriasis, a combined product containing calcipotriol monohydrate and betamethasone dipropionate then vitamin D or vitamin D analogue was statistically significantly better than topical vitamin D or vitamin D analogue for:

  • Investigator’s assessment (clear/nearly clear) for a combined product once daily for 4 weeks then calcipotriol once daily for 4 weeks compared to tacalcitol once daily for 8 weeks; a combined product once daily for 8 weeks then calcipotriol once daily for 4 weeks vs. calcipotriol BD for 12 weeks [2 between-patient studies; 1150 participants; moderate quality evidence]201,298
  • Patient’s assessment (clear/nearly clear) for a combined product once daily for 4 weeks then calcipotriol once daily for 4 weeks compared to tacalcitol once daily for 8 weeks [1 between-patient study; 501 participants; moderate quality evidence]298
  • Percentage change in PASI for a combined product once daily for 4 weeks then calcipotriol once daily weekdays/a combined product once daily at weekends for 8 weeks compared to calcipotriol twice daily for 12 weeks; a combined product once daily for 8 weeks then calcipotriol once daily for 4 weeks vs. calcipotriol twice daily for 12 weeks [1 between-patient study; 972 participants; moderate quality evidence]201
  • Percentage change in PASI for a combined product once daily for 4 weeks then calcipotriol once daily for 4 weeks compared to tacalcitol once daily for 8 weeks [1 between-patient study; 501 participants; moderate quality evidence]298

In people with psoriasis, there were no events with either a combined product containing calcipotriol monohydrate and betamethasone dipropionate then vitamin D or vitamin D analogue or topical vitamin D or vitamin D analogue for:

  • Skin atrophy for a combined product once daily for 4 weeks then calcipotriol once daily weekdays/a combined product once daily at weekends for 8 weeks compared to calcipotriol twice daily for 12 weeks [1 between-patient study; 649 participants; moderate quality evidence]201

In people with psoriasis, there was no statistically significant difference between a combined product containing calcipotriol monohydrate and betamethasone dipropionate then vitamin D or vitamin D analogue and topical vitamin D or vitamin D analogue for:

  • Investigator’s assessment (clear/nearly clear) for a combined product once daily for 4 weeks then calcipotriol once daily weekdays/a combined product once daily at weekends for 8 weeks compared to calcipotriol twice daily for 12 weeks [1 between-patient study; 650 participants; low quality evidence] 201
  • Withdrawal due to adverse events for a combined product once daily for 4 weeks then calcipotriol twice daily for 8 weeks compared to calcipotriol twice daily for 12 weeks [1 between-patient study; 101 participants; very low quality evidence]351
  • Withdrawal due to adverse events for a combined product once daily for 4 weeks then calcipotriol once daily for 4 weeks compared to tacalcitol once daily for 8 weeks [1 between-patient study; 451 participants; very low quality evidence]298
  • Withdrawal due to lack of efficacy for a combined product once daily for 4 weeks then calcipotriol twice daily for 8 weeks compared to calcipotriol twice daily for 12 weeks [1 between-patient study; 100 participants; very low quality evidence]351
  • Withdrawal due to lack of efficacy for a combined product once daily for 4 weeks then calcipotriol once daily for 4 weeks compared to tacalcitol once daily for 8 weeks [1 between-patient study; 445 participants; very low quality evidence]298
  • Skin atrophy for a combined product once daily for 8 weeks then calcipotriol once daily for 4 weeks compared to calcipotriol twice daily for 12 weeks [1 between-patient study; 649 participants; very low quality evidence]201

8.1.12.3. Heterogeneity

  • Not applicable as the studies assessed slightly different comparisons and so were not a combined

8.1.13. Combined product containing vitamin D or vitamin D analogue and potent corticosteroid (calcipotriol plus betamethasone dipropionate) vs. vitamin D or vitamin D analogue (52 weeks maintenance)

This study enrolled patients with plaque psoriasis of at least moderate severity and allowed treatment once daily according to the randomised intervention schedule for up to 52 weeks (52 weeks of the combination product vs 4 weeks of the combination product then 48 weeks with calcipotriol alone vs alternating 4-week periods of treatment with the combination product and calcipotriol alone); however, to accord with clinical practice, topical treatments were only applied when required.

8.1.13.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D and corticosteroid combinationVitamin D or vitamin D analogueRelative (95% CI)Absolute
Investigator's assessment of treatment success (absent, very mild or mild disease) – Combination OD (52 wk) vs. combination OD (4 wk) then calcipotriol OD (48 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriousano serious inconsistencyseriousbseriouscnone80/104 (76.9%)62/89 (69.7%)RR 1.1 (0.93 to 1.31)70 more per 1000 (from 49 fewer to 216 more)⊕○○○
VERY LOW
Investigator's assessment of treatment success (absent, very mild or mild disease) – Combination OD (52 wk) vs. alternating combination OD and calcipotriol OD (52 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriousdno serious inconsistencyseriousbno serious imprecisionnone80/104 (76.9%)78/104 (75%)RR 1.03 (0.88 to 1.2)22 more per 1000 (from 90 fewer to 150 more)⊕⊕○○
LOW
Investigator's assessment of treatment success (absent, very mild or mild disease) -Alternating combination OD and calcipotriol OD (52 wk) vs combination OD (4 wk) then calcipotriol OD (48 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriouseno serious inconsistencyseriousbseriouscnone78/104 (75%)62/89 (69.7%)RR 1.08 (0.9 to 1.28)56 more per 1000 (from 70 fewer to 195 more)⊕○○○
VERY LOW
Skin atrophy -Combination OD (52 wk) vs. combination OD (4 wk) then calcipotriol OD (48 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriousano serious inconsistencyno serious indirectnessvery seriousfnone4/212 (1.9%)2/209 (0.96%)RR 1.97 (0.37 to 10.65)9 more per 1000 (from 6 fewer to 92 more)⊕○○○
VERY LOW
Skin atrophy -Combination OD (52 wk) vs. alternating combination OD and calcipotriol OD (52 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriousdno serious inconsistencyno serious indirectnessvery seriousfnone4/212 (1.9%)1/213 (0.47%)RR 4.02 (0.45 to 35.66)14 more per 1000 (from 3 fewer to 163 more)⊕○○○
VERY LOW
Skin atrophy -Alternating combination OD and calcipotriol OD (52 wk) vs combination OD (4 wk) then calcipotriol OD (48 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriouseno serious inconsistencyno serious indirectnessvery seriousfnone1/213 (0.47%)2/209 (0.96%)RR 0.49 (0.04 to 5.37)5 fewer per 1000 (from 9 fewer to 42 more)⊕○○○
VERY LOW
Withdrawal due to adverse events – Combination OD (52 wk) vs. combination OD (4 wk) then calcipotriol OD (48 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriousano serious inconsistencyno serious indirectnessvery seriousfnone14/162 (8.6%)16/155 (10.3%)RR 0.84 (0.42 to 1.66)17 fewer per 1000 (from 60 fewer to 68 more)⊕○○○
VERY LOW
Withdrawal due to adverse events -Combination OD (52 wk) vs. alternating combination OD and calcipotriol OD (52 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriousdno serious inconsistencyno serious indirectnessvery seriousfnone14/162 (8.6%)11/168 (6.5%)RR 1.32 (0.62 to 2.82)21 more per 1000 (from 25 fewer to 119 more)⊕○○○
VERY LOW
Withdrawal due to adverse events -Alternating combination OD and calcipotriol OD (52 wk) vs combination OD (4 wk) then calcipotriol OD (48 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriouseno serious inconsistencyno serious indirectnessvery seriousfnone11/168 (6.5%)16/155 (10.3%)RR 0.63 (0.3 to 1.32)38 fewer per 1000 (from 72 fewer to 33 more)⊕○○○
VERY LOW
Withdrawal due to lack of efficacy -Combination OD (52 wk) vs. combination OD (4 wk) then calcipotriol OD (48 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriousano serious inconsistencyno serious indirectnessseriouscnone35/183 (19.1%)42/181 (23.2%)RR 0.82 (0.55 to 1.23)42 fewer per 1000 (from 104 fewer to 53 more)⊕⊕○○
LOW
Withdrawal due to lack of efficacy -Combination OD (52 wk) vs. alternating combination OD and calcipotriol OD (52 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriousdno serious inconsistencyno serious indirectnessvery seriousfnone35/183 (19.1%)31/188 (16.5%)RR 1.16 (0.75 to 1.8)26 more per 1000 (from 41 fewer to 132 more)⊕○○○
VERY LOW
Withdrawal due to lack of efficacy -Alternating combination OD and calcipotriol OD (52 wk) vs combination OD (4 wk) then calcipotriol OD (48 wk) (follow-up 52 weeks)
1
Kragballe 2006 (and 2006A)
randomised trialsseriouseno serious inconsistencyno serious indirectnessseriouscnone31/188 (16.5%)42/181 (23.2%)RR 0.71 (0.47 to 1.08)67 fewer per 1000 (from 123 fewer to 19 more)⊕⊕○○
LOW
a

Unclear allocation concealment and blinding; high dropout rate (30% in combination group and 33.5% in calcipotriol group)

b

Definition of success is too broad

c

Confidence interval ranges from clinically important effect to no effect

d

Unclear allocation concealment and blinding; high dropout rate (30% in combination group and 26.3% in alternating group)

e

Unclear allocation concealment and blinding; high dropout rate (26.3% in alternating group and 33.5% in vitamin D or vitamin D analogue group)

f

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

8.1.13.2. Evidence statements

In people with psoriasis, there was no statistically significant difference between the maintenance regimens for 52 weeks maintenance for:

  • Investigator’s assessment of treatment success (absent, very mild or mild disease) at 52 weeks [1 between-patient study; 297 participants; low to very low quality evidence]196
  • Skin atrophy at 52 weeks [1 between-patient study; 634 participants; very low quality evidence]195
  • Withdrawal due to adverse events at 52 weeks [1 between-patient study; 485 participants; very low quality evidence]195,196
  • Withdrawal due to lack of efficacy at 52 weeks [1 between-patient study; 552 participants; low to very low quality evidence]195,196

8.1.13.3. Heterogeneity

  • Not applicable as this study assessed multiple comparisons and combining all results would lead to double counting of data.

8.1.14. Vitamin D or vitamin D analogue vs. dithranol

8.1.14.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D or analogueDithranolRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) -Calcipotriol BD vs. dithranol OD (follow-up 8–12 weeks)
3
Berth Jones 1992 Christensen 1999 Wall 1998
randomised trialsseriousaseriousbno serious indirectnesscseriousdnone278/473 (58.8%)187/435 (43%)RR 1.36 (1.10 to 1.68)155 more per 1000 (from 43 more to 292 more)⊕○○○
VERY LOW
Investigator’s assessment (clear/nearly clear) -Calcitriol BD vs. dithranol OD (follow-up 8 weeks)
1
Hutchinson 2000
randomised trialsseriouseno serious inconsistencyseriousfvery seriousgnone4/60 (6.7%)9/54 (16.7%)RR 0.4 (0.13 to 1.22)100 fewer per 1000 (from 145 fewer to 37 more)⊕○○○
VERY LOW
Patient's assessment (clear/nearly clear) -Calcipotriol BD vs. dithranol OD (follow-up 8–12 weeks)
2
Berth Jones 1992 Wall 1998
randomised trialsserioushno serious inconsistencyno serious indirectnessno serious imprecisionnone273/384 (71.1%)188/358 (52.5%)RR 1.36 (1.21 to 1.53)189 more per 1000 (from 110 more to 278 more)⊕⊕⊕○
MODERATE
% change in PASI -Calcipotriol BD vs. dithranol OD (follow-up 8 weeks; Better indicated by lower values)
1
van de Kerkhof 2006
randomised trialsseriousino serious inconsistencyno serious indirectnessseriousjnone4640-MD 6.6 higher (7.04 lower to 20.24 higher)⊕⊕○○
LOW
Withdrawals due to adverse events -Calcipotriol or calcitriol BD vs. dithranol OD (follow-up 8–12 weeks)
5
Berth Jones 1992
Christensen 1999
Hutchinson 2000
van der Kerkhof 2006
Wall 1998
randomised trialsseriouskno serious inconsistencyno serious indirectnessno serious imprecisionnone22/561 (3.9%)43/524 (8.2%)RR 0.49 (0.3 to 0.79)42 fewer per 1000 (from 17 fewer to 57 fewer)⊕⊕⊕○
MODERATE
Withdrawals due to lack of efficacy -Calcipotriol BD vs. dithranol OD (follow-up 8 weeks)
1
van de Kerkhof 2006
randomised trialsseriousino serious inconsistencyno serious indirectnessseriousjnone7/47 (14.9%)4/49 (8.2%)RR 1.82 (0.57 to 5.83)67 more per 1000 (from 35 fewer to 394 more)⊕⊕○○
LOW
Relapse rate -Calcipotriol BD vs. dithranol OD (8 week post-treatment)
1
Christensen 1999
randomised trialsvery seriouslno serious inconsistencyseriousmseriousnnone50/62 (80.6%)19/33 (57.6%)RR 1.40 (1.02 to 1.92)230 more per 1000 (from 12 more to 530 more)⊕○○○
VERY LOW
Median time to relapse -Calcipotriol BD vs. dithranol OD (follow-up 8 week post-treatment)
1
Christensen 1999
randomised trialsvery seriouslno serious inconsistencyno serious indirectnessvery seriousonone6233-Calcipotriol: 29 days Dithranol: 56 days⊕○○○
VERY LOW
a

3/3 unclear allocation concealment; 2/3 open and 1/3 unclear blinding

b

Heterogeneity was present (I2 = 50%) that could not be explained by pre-defined subgroups (however, all studies showed the same direction of effect)

c

1/3 (2% weighted has strict definition of response - complete clearance)

d

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit in favour of vitamin D or vitamin D analogue to no clinically important difference)

e

Unclear allocation concealment and unblinded; high differential dropout rate (20% vitamin D or vitamin D analogue and 29.6% dithranol)

f

Strict definition of response (complete clearance)

g

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

h

2/2 unclear allocation concealment and unblended

i

Unclear blinding and high differential dropout rate (vitamin D or vitamin D analogue 25.9%; dithranol 13.5%)

j

Confidence interval ranges from clinically significant effect to no effect (default MID = 0.5 × median control group SD = 14.55%)

k

4/5 unclear allocation concealment; 3/5 unblinded and 2/5 unclear blinding; 2/5 (15.5% weighted) high differential dropout rate (one with more dropouts in vitamin D or vitamin D analogue group and one with more in dithranol group)

l

Unclear allocation concealment and blinding; high dropout rate during post-treatment phase (full details not given but appears higher in dithranol group); only includes those who were at least 50% improved and willing to continue; therefore, unclear baseline comparability and fewer in the dithranol group

m

Surrogate outcome for duration of remission

n

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit in favour of dithranol to no clinically important difference)

o

Interpreted from graphical representation

8.1.14.2. Evidence statements

In people with psoriasis, vitamin D or vitamin D analogue was statistically significantly better than dithranol for:

  • Investigator’s assessment (clear/nearly clear) at 8–12 weeks for calcipotriol twice daily compared to dithranol once daily [3 between-patient studies; 908 participants; very low quality evidence]29,58,417
  • Patient’s assessment (clear/nearly clear) at 8–12 weeks for calcipotriol twice daily compared to dithranol once daily [2 between-patient studies; 742 participants; moderate quality evidence]29,417
  • Withdrawals due to adverse events at 8–12 weeks for calcipotriol or calcitriol twice daily compared to dithranol once daily [5 between-patient studies; 1085 participants; moderate quality evidence] 29,58,156,410,417

In people with psoriasis, dithranol was statistically significantly better than vitamin D or vitamin D analogue for:

  • Relapse rate at 8 weeks post treatment for calcipotriol twice daily compared to dithranol once daily [1 between-patient study; 95 participants; very low quality evidence]58

In people with psoriasis, there was no statistically significant difference between dithranol and vitamin D or vitamin D analogue for:

  • Investigator’s assessment (clear/nearly clear) at 8 weeks for calcitriol twice daily compared to dithranol once daily [1 between-patient study; 114 participants; very low quality evidence]156
  • Percentage change in PASI at 8 weeks for calcipotriol twice daily compared to dithranol once daily [1 between-patient study; 86 participants; low quality evidence]410
  • Withdrawals due to lack of efficacy at 8 weeks for calcipotriol twice daily compared to dithranol once daily [1 between-patient study; 96 participants; low quality evidence] 410
Evidence statement for individual study where no statistical analysis could be performed

In people with psoriasis, dithranol was better than vitamin D or vitamin D analogue for:

  • Median time to relapse for a maximum follow-up of at 8 weeks post-treatment among those who had achieved remission with calcipotriol twice daily compared to dithranol once daily [1 between-patient study; 95 participants; very low quality evidence]58

8.1.14.3. Heterogeneity

  • For the outcome of investigator’s assessment of achieving clear/nearly clear status heterogeneity was present. The heterogeneity was greatly reduced by separating into subgroups based on the specific vitamin D or vitamin D analogue used; suggesting that calcitriol may be less effective than dithranol but calcipotriol may be more effective. However, there was still some heterogeneity among the studies using calcipotriol, although all showed the same direction of effect.
  • There was no significant heterogeneity for the remaining outcomes.

8.1.15. Vitamin D or vitamin D analogue vs. coal tar

8.1.15.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D or vitamin D analogueCoal tarRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) -Calcipotriol BD vs 15% coal tar solution in aqueous cream OD (follow-up 6 weeks)
1
Tham 1994
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone13/27 (48.1%)3/27 (11.1%)RR 4.33 (1.39 to 13.5)370 more per 1000 (from 43 more to 1000 more)⊕⊕⊕○
MODERATE
Investigator’s assessment (clear/nearly clear) -Calcipotriol BD vs. coal tar polytherapy (coal tar 5%/allantoin 2%/hydrocortisone cream 0.5%) BD (follow-up 8 weeks)
1
Pinheiro 1997
randomised trialsseriousbno serious inconsistencyno serious indirectnessseriouscnone47/65 (72.3%)28/57 (49.1%)RR 1.47 (1.09 to 1.99)231 more per 1000 (from 44 more to 486 more)⊕⊕○○
LOW
Investigator’s assessment (clear/nearly clear) -Calcipotriol BD vs. coal tar solution (liquor carbonis distillate (LCD 15%, equivalent to 2.3% coal tar) BD (follow-up 12 weeks)
1
Alora-Palli 2010
randomised trialsvery seriousdno serious inconsistencyno serious indirectnessseriousenone6/28 (21.4%)14/27 (51.9%)RR 0.41 (0.19 to 0.92)306 fewer per 1000 (from 41 fewer to 420 fewer)⊕○○○
VERY LOW
% change in PASI -Calcipotriol BD vs 15% coal tar solution in aqueous cream OD (follow-up 6 weeks; Better indicated by lower values)
1
Tham 1994
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone2727-MD 38.9 lower (50.95 to 26.85 lower)⊕⊕⊕○
MODERATE
% change in PASI - Calcipotriol BD vs. coal tar solution (liquor carbonis distillate (LCD 15%, equivalent to 2.3% coal tar) BD (follow-up 12 weeks; Better indicated by lower values)
1
Alora-Palli 2010
randomised trialsvery seriousdno serious inconsistencyno serious indirectnessno serious imprecisionnone2827-MD 21.7 higher (4.2 to 39.2 higher)⊕⊕○○
LOW
Relapse rate (6 weeks post-treatment) - Calcipotriol BD vs. coal tar solution (liquor carbonis distillate (LCD 15%, equivalent to 2.3% coal tar) BD
1
Alora-Palli 2010
randomised trialsvery seriousfno serious inconsistencyseriousgno serious imprecisionnone7/9 (77.8%)4/16 (25%)RR 3.11 (1.24 to 7.79)527 more per 1000 (from 85 more to 1000 more)⊕○○○
VERY LOW
Withdrawals due to adverse events - Calcipotriol BD vs 15% coal tar solution in aqueous cream OD (follow-up 6 weeks)
1
Tham 1994
randomised trialsseriousano serious inconsistencyno serious indirectnessvery serioushnone1/25 (4%)0/25 (0%)RR 3 (0.13 to 70.3)-⊕○○○
VERY LOW
Withdrawals due to adverse events - Calcipotriol BD vs. coal tar polytherapy (coal tar 5%/allantoin 2%/hydrocortisone cream 0.5%) BD (follow-up 8 weeks)
1
Pinheiro 1997
randomised trialsseriousbno serious inconsistencyno serious indirectnessvery serioushnone1/62 (1.6%)3/54 (5.6%)RR 0.29 (0.03 to 2.71)39 fewer per 1000 (from 54 fewer to 95 more)⊕○○○
VERY LOW
Withdrawals due to adverse events - Calcipotriol BD vs. coal tar solution (liquor carbonis distillate (LCD 15%, equivalent to 2.3% coal tar) BD (follow-up 12 weeks)
1
Alora-Palli 2010
randomised trialsvery seriousdno serious inconsistencyno serious indirectnessno serious imprecisionnone0/28 (0%)0/27 (0%)not poolednot pooled⊕⊕○○
LOW
a

Unclear allocation concealment and blinding

b

Unclear allocation concealment and unblended

c

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit of vitamin D or vitamin D analogues to no clinically important difference)

d

Unclear allocation concealment, single blind (investigator) and high differential dropout rate (16.7% in tar and 26.7% in calcipotriol group during treatment phase)

e

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit of coal tar to no clinically important difference)

f

Unclear allocation concealment, single blind (investigator) and high differential dropout rate (16.7% in tar and 26.7% in calcipotriol group during treatment phase); also only include those who achieved a PASI50; therefore, unclear baseline comparability and fewer in the calcipotriol group

g

Surrogate outcome for duration of remission

h

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

8.1.15.2. Evidence statements

In people with psoriasis, vitamin D or vitamin D analogue treatment was statistically significantly better than coal tar for:

  • Investigator’s assessment (clear/nearly clear) at 6–8 weeks for calcipotriol twice daily compared to 15% coal tar solution in aqueous cream once daily; calcipotriol twice daily compared to coal tar polytherapy (coal tar 5%/allantoin 2%/hydrocortisone cream 0.5%) twice daily [2 studies (1 within- and 1 between-patient); 149 participants (176 randomised units); low to moderate quality evidence]313,400
  • Percentage change in PASI at 6 weeks for calcipotriol twice daily compared to 15% coal tar solution in aqueous cream once daily [1 within-patient study; 27 participants (54 randomised units); moderate quality evidence]400

In people with psoriasis, coal tar was statistically significantly better than vitamin D or vitamin D analogue for:

  • Investigator’s assessment (clear/nearly clear) at 12 weeks for calcipotriol twice daily compared to coal tar solution (liquor carbonis distillate (LCD 15%, equivalent to 2.3% coal tar) twice daily [1 between-patient study; 55 participants; very low quality evidence]15
  • Percentage change in PASI at 12 weeks for calcipotriol twice daily compared to coal tar solution (liquor carbonis distillate (LCD 15%, equivalent to 2.3% coal tar) twice daily [1 between-patient study; 55 participants; low quality evidence]15
  • Relapse rate at 6 weeks post-treatment for calcipotriol twice daily compared to coal tar solution (liquor carbonis distillate (LCD 15%, equivalent to 2.3% coal tar) twice daily [1 between-patient study; 25 participants; very low quality evidence]15

In people with psoriasis, there were no events with either vitamin D or vitamin D analogue or coal tar for:

  • Withdrawals due to adverse events at 12 weeks for calcipotriol twice daily compared to coal tar solution (liquor carbonis distillate (LCD 15%, equivalent to 2.3% coal tar) twice daily [1 between-patient study; 55 participants; low quality evidence]15

In people with psoriasis, there was no statistically significant difference between vitamin D or vitamin D analogue and coal tar for:

  • Withdrawals due to adverse events at 6 weeks for calcipotriol twice daily compared to 15% coal tar solution in aqueous cream once daily [1 within patient study; 25 participants (50 randomised units); very low quality evidence]400
  • Withdrawals due to adverse events at 8 weeks for calcipotriol twice daily compared to coal tar polytherapy (coal tar 5%/allantoin 2%/hydrocortisone cream 0.5%) twice daily [1 between-patient study; 116 participants; very low quality evidence]313

8.1.15.3. Heterogeneity

  • Heterogeneity was present for all outcomes. The heterogeneity was removed by separating into subgroups based on treatment duration. However, it is also possible that the coal tar formulation caused the heterogeneity, although this was thought to be clinically less likely to be the source of the inconsistency.

8.1.16. Vitamin D or vitamin D analogue once daily compared to vitamin D or vitamin D twice daily

8.1.16.1. Evidence profile

Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin DODVitamin DBDRelative (95% CI)Absolute
Investigator's assessment (clear/nearly clear) – calcipotriol (follow-up 8 weeks)
1
Kragballe 1998
randomised trialsseriousano serious inconsistencyno serious indirectnessseriousbnone49/172 (28.5%)69/172 (40.1%)RR 0.71 (0.53 to 0.96)116 fewer per 1000 (from 16 fewer to 189 fewer)⊕⊕○○
LOW
Patient's assessment (clear/nearly clear) – calcipotriol (follow-up 8 weeks)
1
Kragballe 1998
randomised trialsseriousano serious inconsistencyno serious indirectnessseriousbNone46/172 (26.7%)69/172 (40.1%)RR 0.67 (0.49 to 0.91)132 fewer per 1000 (from 36 fewer to 205 fewer)⊕⊕○○
LOW
Withdrawals due to adverse events – calcipotriol (follow-up 8 weeks)
1
Kragballe 1998
randomised trialsseriousano serious inconsistencyno serious indirectnessvery seriouscNone8/174 (4.6%)6/174 (3.4%)RR 1.33 (0.47 to 3.76)11 more per 1000 (from 18 fewer to 95 more)⊕○○○
VERY LOW
Withdrawals due to lack of efficacy – calcipotriol (follow-up 8 weeks)
1
Kragballe 1998
randomised trialsseriousano serious inconsistencyno serious indirectnessvery seriouscNone2/174 (1.1%)3/174 (1.7%)RR 0.67 (0.11 to 3.94)6 fewer per 1000 (from 15 fewer to 51 more)⊕○○○
VERY LOW
a

Unclear allocation concealment and blinding

b

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit in favour of twice daily application to no clinically important difference)

c

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

8.1.16.2. Evidence statements

In people with psoriasis, calcipotriol twice daily was statistically significantly better than calcipotriol once daily for:

  • Investigator’s assessment (clear/nearly clear) at 8 weeks [1 within-patient study; 344 participants; low quality evidence]197
  • Patient’s assessment (clear/nearly clear) at 8 weeks [1 within-patient study; 344 participants; low quality evidence]197

In people with psoriasis, there was no statistically significant difference between calcipotriol once daily and calcipotriol twice daily for:

  • Withdrawal due to adverse events at 8 weeks [1 within-patient study; 348 participants; very low quality evidence]197
  • Withdrawal due to lack of efficacy at 8 weeks [1 within-patient study; 348 participants; very low quality evidence]197

8.1.16.3. Heterogeneity

  • Not applicable as only one study was available for this comparison

8.1.17. Time to remission or maximum effect for trunk or limb psoriasis

8.1.17.1. Vitamin D or vitamin D analogues

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D or vitamin D analogue
Time-to-remission (marked improvement or clearance (follow-up 1–8 weeks)
1
Highton 1995
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneCalcipotriol BD
124
Patients achieving marked improvement or clearance⊕⊕○○
LOW
Week 19.6%
Week 227.8%
Week 454.2%
Week 665.1%
Week 8/EOT69.8%
Time-to-remission (clear/nearly clear; follow-up 4–8 weeks)
1
Fleming2010A
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneCalcipotriol OD
79
Clear/nearly clear (investigator’s static assessment)⊕⊕○○
LOW
Week 426 (16.0%)
Week 844 (27.2%)
Time-to-remission (clear/nearly clear; follow-up 4–8 weeks)
1
Langley 2011A
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneTacalcitol OD
184
Clear/nearly clear (investigator’s static assessment)⊕⊕○○
LOW
Week 412 (6.5%)
Week 833 (17.9%)
Clear/nearly clear (patient’s static assessment)
Week 421/175 (12.0%)
Week 835/163 (21.5%)
Time-to-maximum response (change in PASI; follow-up 2–6 weeks)
1
Cunliffe 1992
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneCalcipotriol BD
201
Mean (SD) change in PASI from baseline (mean at baseline = 8.67)⊕⊕○○
LOW
Week 23.19 (3.61)
Week 44.37 (4.70)
Week 65.5 (9.54)
Time-to-maximum response (change in PASI; follow-up 2–4 weeks)
1
Dubertret 1992
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneCalcipotriol BD
65
Mean (SD) PASI during initial 4-week randomised treatment phase⊕⊕○○
LOW
Mean baseline PASI (n=65)14.2 ± 7.5
After 2 weeks (n=62)
Mean PASI8.6 ± 7.5
% change from baseline41.2 ± 25.7
After 4 weeks (n=60)
Mean PASI6.3 ± 6.5
% change from baseline58.6 ± 31.7
Time-to-maximum response (change in PASI; follow-up 2–12 weeks)
1
Saraceno 2007
observational studiesano serious risk of biascno serious inconsistencyno serious indirectnessno serious imprecisionnoneCalcipotriol BD
75
Mean PASI (SD)⊕⊕○○
LOW
Baseline9.11 (4.09)
2 weeks5.47 (3.47)
4 weeks4.07 (3.33)
8 weeks3.45 (3.77)
12 weeks3.04 (3.76)
Time-to-maximum response (% change in PASI; follow-up 2–4 weeks)
1
Ortonne 2004
observational studiesano serious risk of biasdno serious inconsistencyno serious indirectnessno serious imprecisionnoneTacalcitol OD
252
Mean % reduction in PASI score from baseline⊕⊕○○
LOW
2 weeks24.5%
4 weeks33.3%
Time-to-maximum response (% change in PASI; follow-up 4–8 weeks)
1
Langley 2011A
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneTacalcitol OD
184
% change in PASI⊕⊕○○
LOW
week 4−37.3
week 8−41.9
Time-to-maximum response (% change in mPASI [0.64.8]; follow-up 4–12 weeks)
1
Alora-Palli 2010
observational studiesano serious risk of biaseno serious inconsistencyno serious indirectnessno serious imprecisionnoneCalcipotriol BD
28
% change in PASI from baseline⊕⊕○○
LOW
Baseline7.07
4 weeks5.09 (−30.2%)
8 weeks4.71 (−34.2%)
12 weeks4.66 (−36.5%)
Time-to-maximum response (% change in PASI; follow-up 2–6 weeks)
1
Tham 1994
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneCalcipotriol BD
27
Mean PASI (italics) and % change in PASI score from baseline⊕⊕○○
LOW
Baseline6.6±4.9
2 weeks4.1±3.4
−36.9±25.0%
4 weeks2.8±2.2
−57.5±19.4%
6 weeks2.0±2.1
−69.8±20.4%
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Unclear allocation concealment may have biased patient selection for this intervention and there was a high rate of dropout (38.7%)

d

Unclear allocation concealment may have biased patient selection for this intervention and there was a high rate of dropout (20.2%)

e

Unclear allocation concealment may have biased patient selection for this intervention and there was a high rate of dropout (26.7%)

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for vitamin D or vitamin D analogues (no statistical analysis could be performed).

In people with psoriasis, the time to remission when using vitamin D or vitamin D analogues varied between studies:

  • Proportion achieving remission by 8 weeks ranged from 11.4 to 69.8% [3 studies; 387 participants; low quality evidence]102,148,208
  • The continued increase in responders between 4 and 8 weeks ranged from 7.6–15.6% [3 studies; 387 participants; low quality evidence]102,148,208
  • The continued increase in responders between 6 and 8 weeks was 4.7% [1 study; 124 participants; low quality evidence]148
  • Of those who achieved remission by the end of the trial at 8 weeks, 33.3–77.7% had responded by week 4 and 93.3% by week 6 on calcipotriol; but just 36.4% of those who achieved remission by the end of the trial had responded by week 4 on tacalcitol [3 studies; 387 participants; low quality evidence]102,148,208
  • The decrease in PASI from 2–4 weeks ranged from 1.18–2.4 points [4 studies; 368 participants; low quality evidence]66,81,351,400
  • The continued decrease in PASI from 4–6 weeks ranged from 0.8–1.13 points [2 studies; 228 participants; low quality evidence]66,400
  • The continued decrease in PASI from 8–12 weeks ranged from 0.05–0.41 points [2 studies; 103 participants; low quality evidence]15,351
  • The % decrease in PASI from 2–4 weeks ranged from 8.8–20.6% [5 studies; 620 participants; low quality evidence]66,81,298,351,400
  • The % decrease in PASI from 4 to 6 or 8 weeks ranged from 4.0–13.0% and from 8–12 weeks from 2.3–4.5% [5 studies; 515 participants; low quality evidence]15,66,208,351,400
  • The % decrease in PASI from 8–12 weeks ranged from 0.7–4.5% [2 studies; 103 participants; low quality evidence]15,351
Summary

The evidence suggests that maximum response is not achieved in all patients by 8–12 weeks, with the response rate still increasing slightly at this time point, although the most rapid improvement was seen over the first 2–4 weeks, particularly for twice daily application.

8.1.17.2. Potent corticosteroids

Evidence profiles
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsPotent corticosteroid
Time-to- clearance or near clearance (follow-up 4–8 weeks)
1
Fleming2010A
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneBetamethasone dipropionate OD
83
Clear/nearly clear (investigator’s static assessment)⊕⊕○○
LOW
Week 48 (9.6%)
Week 814 (16.9%)
Time-to-marked improvement or clearance (follow-up 8–22 days)
1
Medansky 1987
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneMometasone furoate OD
58
Patients achieving marked improvement or clearance⊕⊕○○
LOW
8 days4/58 (6.9%)
15 days12/55 (21.8%)
22 days18/50 (36.0%)
Time-to-excellent or good improvement (follow-up 7–21 days)
1
Sears 1997
observational studiesano serious risk of biascno serious inconsistencyserious indirectnessdno serious imprecisionnoneHydrocortisone buteprate BD
84
Patients achieving excellent or good improvement⊕○○○
VERY LOW
Day 7:15/84 (17.9%)
Day 14:24/84 (28.2%)
Day 21:32/78 (41.3%)
Mean time to remission (IAGI – clear, excellent or good) (follow-up 4 weeks)
1
Olsen 1996 – study A
observational studiesano serious risk of biasbno serious inconsistencyseriousdno serious imprecisionseriouseFluticasone propionate BD
88
Investigator’s assessment⊕○○○
VERY LOW
Week 1
Clear0
Excellent/good55%
Week 2
Clear4%
Excellent/good60%
Week 3
Clear4%
Excellent/good65%
Week 4
Clear11%
Excellent/good60%
Mean time to remission (IAGI – clear, excellent or good) (follow-up 4 weeks)
1
Olsen 1996 – study B
observational studiesano serious risk of biasbno serious inconsistencyseriousdno serious imprecisionseriouseFluticasone propionate BD
105
Investigator’s assessment⊕○○○
VERY LOW
Week 1
Clear0
Excellent/good29%
Week 2
Clear0
Excellent/good50%
Week 3
Clear0
Excellent/good65%
Week 4
Clear3%
Excellent/good66%
Time-to-maximum response (% change in PASI; follow-up 2–6 weeks)
1
Thawornchaisit 2007
observational studiesano serious risk of biascno serious inconsistencyno serious indirectnessno serious imprecisionnoneBetamethasone valerate BD
30
Mean PASI and % change in PASI score from baseline⊕⊕○○
LOW
2 weeks12.95±3.4
−27.23±10.6%
4 weeks8.68±3.8
−51.41±18.2%
6 weeks5.52±4.5
−69.36±23.3%
Time-to-maximum response (change in PASI; follow-up 2–6 weeks)
1
Cunliffe 1992
observational studiesano serious risk of biascno serious inconsistencyno serious indirectnessno serious imprecisionnoneBetamethasone valerate BD

200
Mean (SD) change in PASI from baseline⊕⊕○○
LOW
Mean at baseline9.35
2 weeks3.39 (2.16)
4 weeks4.50 (5.33)
6 weeks5.32 (6.06)
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention and there was a high rate of dropout (21.5%)

c

Unclear allocation concealment may have biased patient selection for this intervention

d

Incorrect definition of response

e

Note that only percentages of responders are available and it is unclear whether the number of participants were assessed at each time point

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for potent corticosteroids (no statistical analysis could be performed).

In people with psoriasis, the time to remission when using potent corticosteroids varied between studies:

  • Proportion achieving remission by 3 weeks ranged from 36.0–41.3% on mometasone furoate or hydrocortisone buteprate [2 studies; 142 participants; low to very low quality evidence]246,360
  • Proportion achieving remission by 4 weeks on fluticasone propionate ranged from 69–71% [2 studies; 793 participants; very low quality evidence]291
  • Proportion achieving remission by 8 weeks on betamethasone dipropionate was 16.9% [1 study; 83 participants; low quality evidence]102
  • The continued increase in responders on mometasone furoate or hydrocortisone buteprate between 2 and 3 weeks ranged from 13.1–14.2%, meaning that 66.7 to 75.0% of those who responded during the trial had achieved remission by 2 weeks [2 studies; 142 participants; low to very low quality evidence]246,360
  • The continued increase in responders between 4–8 weeks of treatment on betamethasone dipropionate, was 7.3% [1 study; 83 participants; low quality evidence]102
  • The continued increase in responders between 3–4 weeks of treatment on fluticasone propionate, ranged from 2–4% [2 studies; 193 participants; very low quality evidence] 291
  • Of those who achieved remission by the end of the trial at 3 weeks, 66.7 to 75.0% had responded by week 2 on mometasone furoate or hydrocortisone buteprate [2 studies; 142 participants; low to very low quality evidence]246,360
  • Of those who achieved remission by the end of the trial at 4 weeks, 72.5–83.1% had responded by week 2 and 89.6–94.2% by week 3 on fluticasone propionate [2 studies; 193 participants; very low quality evidence]291
  • Of those who achieved remission by the end of the trial at 8 weeks on betamethasone dipropionate, 57.1% had responded by week 4 [1 study; 83 participants; low quality evidence]102
  • The continued decrease in PASI on betamethasone valerate from 4–6 weeks ranged from 0.82–3.16 points/8.8–17.95% [2 studies; 230 participants; low quality evidence]66,401
Summary

The evidence suggests that maximum response is not achieved in all patients by 6–8 weeks, with the response rate still increasing slightly at this time point, although the most rapid improvement was seen over the first 2–4 weeks, particularly for twice daily application.

8.1.17.3. Very potent corticosteroids

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVery potent corticosteroid
Mean time to maximum response (global severity score) (follow-up 4 weeks)
1
Decroix 2004
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessvery seriouscnoneClobetasol propionate OD

189
Mean global severity score over time shows that maximum effect is not achieved by week 4 (gradual improvement still apparent)⊕○○○
VERY LOW
Mean time to maximum response (TSS) (follow-up 4 weeks)
1
Lowe 2005
observational studiesano serious risk of biasbno serious inconsistencyseriousdvery seriouscnoneClobetasol propionate BD

162
Mean % change in TSS over time shows that maximum effect is not achieved by week 4⊕○○○
VERY LOW
Mean time to maximum response (TSS) (follow-up 4 weeks)
1
Beutner 2006
observational studiesano serious risk of biasbno serious inconsistencyseriousdvery seriouscnoneClobetasol propionate BD

25
Mean TSS over time shows that maximum effect is not achieved by week 4 (gradual improvement still apparent)⊕○○○
VERY LOW
Mean time to maximum response (TSS) (follow-up 2 weeks)
1
Lebwohl 2002
observational studiesano serious risk of biasbno serious inconsistencyseriousdvery seriouscnoneClobetasol propionate BD

61
Mean TSS over time shows that maximum effect is not achieved by week 2⊕○○○
VERY LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Interpreted from graphical representation

d

Incorrect outcome measure

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for very potent corticosteroids (no statistical analysis could be performed).

In people with psoriasis, the time to remission or maximum response when using very potent corticosteroids varied between studies:

  • Mean change in global severity score showed that a maximum effect was not reached by week 4 [4 studies; 437 participants; very low quality evidence]32,73,216,227
  • Mean change (or % change) in TSS showed that a maximum effect was not reached by week 2 or 4 [4 studies; 437 participants; very low quality evidence]32,73,216,227
Summary

The evidence suggests that maximum response is not achieved in all patients by 2 or 4 weeks, with the response rate still increasing slightly at this time point. However, the most rapid effect is seen over the first 2 weeks.

8.1.17.4. Combined product containing vitamin D or vitamin D analogue and potent corticosteroid (calcipotriol plus betamethasone dipropionate)

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCombination
Time-to-clear/nearly clear (investigator’s assessment; follow-up 4–8 weeks)
1
Langley 2011A
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone183Clear/nearly clear (IGA)⊕⊕○○
LOW
Week 434 (18.6%)
Week 873 (39.9%)
Time-to-clear/nearly clear (investigator’s assessment; follow-up 4–8 weeks)
1
Fleming 2010A
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone162Clear/nearly clear (IGA)⊕⊕○○
LOW
Week 426 (16.0%)
Week 844 (27.2%)
Time-to-clear/nearly clear (patient’s assessment; follow-up 4–8 weeks)
1
Langley 2011A
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone183Clear/nearly clear (patient rating)⊕⊕○○
LOW
Week 452/175 (29.7%)
Week 869/171 (40.4%)
Time-to-maximum effect (% change in PASI; follow-up 4–8 weeks)
1
Langley 2011A
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone183% change in PASI⊕⊕○○
LOW
Week 4−53.1
Week 8−57.0
Time-to-maximum effect (% change in PASI; follow-up 2–4 weeks)
1
Ortonne 2004
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone249Mean % reduction in PASI score from baseline⊕⊕○○
LOW
2 weeks50.5%
4 weeks65.0%
Time-to-maximum effect (change in PASI; follow-up 2–4 weeks)
1
Saraceno 2007
observational studiesano serious risk of biascno serious inconsistencyno serious indirectnessno serious imprecisionnone75Mean PASI (SD)⊕⊕○○
LOW
Baseline9.49 (5.39)
2 weeks3.81 (3.27)
4 weeks2.50 (2.50)
Mean time to maximum response (IAGI) (follow-up 52 weeks)
1
Kragballe 2006
observational studiesano serious risk of biasdno serious inconsistencyno serious indirectnessvery seriousenone212Graph of % satisfactory responses by investigator assessment shows that maximum response is achieved by 12 weeks⊕○○○
VERY LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Unclear allocation concealment may have biased patient selection for this intervention and there was a high rate of dropout (33.3%)

d

Unclear allocation concealment may have biased patient selection for this intervention and there was a high rate of dropout (30.2%)

e

Interpreted from graphical representation

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for a combined product containing vitamin D or vitamin D analogue and potent corticosteroid (calcipotriol plus betamethasone dipropionate; no statistical analysis could be performed).

In people with psoriasis, the time to remission when using a combined product containing vitamin D or vitamin D analogue and potent corticosteroid (calcipotriol plus betamethasone dipropionate) varied between studies:

  • Proportion achieving remission (investigator’s or patient’s assessment) by 8 weeks ranged from 27.2 to 40.4% [2 studies; 345 participants; low quality evidence]102,208
  • The continued increase in responders (investigator’s or patient’s assessment) between 4 and 8 weeks ranged from 10.7–21.3% [2 studies; 345 participants; low quality evidence]102,208
  • Of those who achieved remission by the end of the trial, 46.6–59.1% had responded by week 4 based on Investigator’s assessment, but the figure was 75.4% based in patient’s assessment [2 studies; 345 participants; low quality evidence]102,208
  • The decrease in PASI from 2–4 weeks ranged from 14.5–14.7% [2 studies; 324 participants; low quality evidence]298,351
  • The decrease in PASI from 4–8 weeks was 3.9% [1 study; 183 participants; low quality evidence]208
  • Graphical representation of longer-term data demonstrated that the maximum rate of satisfactory responses based on investigator assessment score was achieved by 12 weeks based on once daily administration as needed, with negligible further improvement up to 12 months [1 study; 212 participants; very low quality evidence]196
Summary

The evidence suggests that maximum response is not achieved in all patients by 4–8 weeks, with the response rate still increasing slightly at this time point. One study196 suggested that 12 weeks may represent the time at which maximum achievement of satisfactory response is achieved based on once daily administration of a combined product containing calcipotriol monohydrate and betamethasone dipropionate as needed, although there was only minimal improvement after 4 weeks.

8.1.17.5. Concurrent potent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening)

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsConcurrent
Time-to-maximum response (change in PASI; follow-up 4 weeks)
1
Ruzika 1998
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessvery seriouscnoneCalcipotriol + betamethasone valerate

78
Based on PASI score over time maximum effect was not reached by 4 weeks of concurrent treatment in the randomised phase (following 2 weeks of calcipotriol treatment)⊕○○○
VERY LOW
Time-to-maximum response (change and % change in PASI; follow-up 8 weeks)
1
Kragballe 1998
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessvery seriouscnoneCalcipotriol + betamethasone valerate

176
Based on change in PASI (and % change in PASI) maximum treatment effect had not been reached by 8 weeks⊕○○○
VERY LOW
Time-to-maximum response (change in PASI; follow-up 4 weeks)
1
Salmhofer 2000
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessvery seriouscnoneCalcipotriol + diflucortolone valerate

63
Based on mean PASI, rapid improvement was seen over first 2 weeks but continued gradual improvement seen up to 4 weeks (maximum effect not reached)⊕○○○
VERY LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Interpreted from graphical representation

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for concurrent potent corticosteroid and vitamin D or vitamin D analogues (one applied in the morning and one in the evening; no statistical analysis could be performed).

In people with psoriasis, the time to remission when using concurrent potent corticosteroid and vitamin D or vitamin D analogues (one applied in the morning and one in the evening):

  • Mean change (or % change) in PASI showed that a maximum effect was not reached by week 4 or 8 [3 studies; 317 participants; very low quality evidence]197,344,346
Summary

The evidence suggests that maximum response is not achieved in all patients by 4–8 weeks, with the response rate still increasing at this time point based on PASI score.

8.1.17.6. Coal tar

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCoal tar
Mean time to maximum response (% change in PASI) (follow-up 6 weeks)
1
Thawornchaisit 2007
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone10% liquor carbonis detergens

28
Mean PASI and % change in PASI score from baseline⊕⊕○○
LOW
2 weeks14.83±3.0
−13.56±8.5%
4 weeks12.31±3.3
−28.18±16.5%
6 weeks10.60±4.1
−38.39±21.1%
Mean time to maximum response (% change in PASI) (follow-up 6 weeks)
1
Tham 1994
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneLiquor picis carbonis

27
Mean PASI and % change in PASI score from baseline⊕⊕○○
LOW
Baseline12.95±3.4
2 weeks5.9±4.5
−9.4±15.9%
4 weeks5.1±4.2
−22.3±24.2%
6 weeks4.5±3.6
−30.9±24.6%
Mean time to maximum response (% change in mPASI [0–64.8]) (follow-up 12 weeks)
1
Alora-Palli 2010
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneLiquor carbonis detergens

27
% change in PASI (0–64.8) from baseline⊕⊕○○
LOW
Baseline7.3
4 weeks4.69 (−35.4%)
8 weeks3.70 (−48.9%)
12 weeks3.24 (−58.2%)
Mean time to maximum response (TSS; follow-up 8 weeks)
1
Pinheiro 1997
observational studiesano serious risk of biasbno serious inconsistencyvery seriouscvery seriousdnoneAlphosyl HC

65
The maximum response based on mean TSS was seen at 4 weeks, with no further improvement up to 8 weeks⊕⊕○○
LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Incorrect outcome measure

d

Interpreted from graphical representation

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for coal tar (no statistical analysis could be performed).

In people with psoriasis, the time to remission when using coal tar varied between studies:

  • The continued % decrease in PASI from 2–4 weeks ranged from 12.9–14.62% (0.8–2.52 PASI points) [2 studies; 55 participants; low quality evidence]400,401
  • The continued % decrease in PASI from 4 to 6 or 8 weeks ranged from 8.6–13.5% (0.6–1.71 PASI points) [3 studies; 82 participants; low quality evidence]15,400,401
  • The decrease in PASI from 8–12 weeks was 9.3% (0.46 PASI points) [1 study; 27 participants; low quality evidence]15
  • Mean change in TSS demonstrated that the maximum response was achieved by 4 weeks, with negligible further improvement up to 8 weeks [1 study; 65 participants; very low quality evidence]313
Summary

The evidence suggests that maximum response to LCD or LPC based on PASI is not achieved in all patients by 6–12 weeks, although the continued absolute change in PASI is small. However, based on TSS, maximum response was seen at 4 weeks when using the Alphosyl HC formulation.

8.1.17.7. Dithranol

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsDithranol
Mean time to maximum response (change in global improvement score; follow-up 8 weeks)
1
Hutchinson 2000
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessvery seriouscnone0.25–2.0% cream (for 30 mins)

60
Based on change in global improvement score over time the maximum treatment effect had not been reached by 8 wks, although the most rapid improvement was seen over the first 4 weeks, with much more gradual reduction between 4–8 wk⊕○○○
VERY LOW
Mean time to maximum response (mean PASI) (follow-up 8 weeks)
1
Hutchinson 2000
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessvery seriouscnone0.25–2.0% cream (for 30 mins)

60
Based on mean PASI, maximum effect appeared to be reached between weeks 6 and 8⊕○○○
VERY LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention and there was a high rate of dropout (29.6%)

c

Interpreted from graphical representation

d

Unclear allocation concealment may have biased patient selection for this intervention

e

Incorrect outcome measure

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for dithranol (no statistical analysis could be performed).

In people with psoriasis, the time to remission when using dithranol was as follows:

  • Mean change in global improvement showed that a maximum effect was not reached by week 8 [1 study; 60 participants; very low quality evidence]156
  • Mean change in PASI showed that a maximum effect was reached by week 6–8 [1 study; 60 participants; very low quality evidence]156
Summary

The evidence suggests that maximum response to dithranol is achieved by 8 weeks of treatment based on change in PASI, but not when assessed using a global improvement score, although even on this outcome the most rapid and pronounced improvement was seen over the first 4 weeks156.

8.1.17.8. Tazarotene

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsTazarotene
Time-to-remission (at least good improvement; follow-up 12 weeks)
1
Weinstein 1997
observational studiesano serious risk of biasbno serious inconsistencyseriouscvery seriousdnone211Based on graphical representation of the % with good or excellent improvement or clearing the maximum response rate had not been reached by 12 weeks⊕○○○
VERY LOW
Time-to-remission (none, minimal or mild disease; follow-up 12 weeks)
1
Weinstein 2003
observational studiesano serious risk of biasbno serious inconsistencyseriouscvery seriousdnone439Based on graphical representation of the % with none, minimal or mild disease the maximum response rate had not been reached by 12 weeks⊕○○○
VERY LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Incorrect definition of response

d

Interpreted for graphical representation

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for tazarotene (no statistical analysis could be performed).

In people with psoriasis, the time to remission when using tazarotene was as follows:

  • Proportion achieving remission had not reached a maximum by 12 weeks [2 studies; 650 participants; very low quality evidence]419,420
Summary

The evidence suggests that maximum proportion achieving remission was not achieved by 12 weeks.

8.1.18. Network meta-analysis for trunk or limb psoriasis

Based on the results of conventional meta-analyses of direct evidence alone, it can be difficult to determine which intervention is most effective in the treatment of chronic plaque psoriasis. The challenge of interpretation arises for two reasons:

  • Some pairs of alternative strategies have not been directly compared in a randomised controlled trial (for example, concurrent vitamin D or vitamin D and potent corticosteroid [one applied in the morning and one in the evening] vs a combined product containing vitamin D or vitamin D analogue and potent corticosteroid)
  • There are frequently multiple overlapping comparisons (for example vitamin D or vitamin D analogue vs potent corticosteroid, vitamin D or vitamin D analogue vs a combined product containing vitamin D or vitamin D analogue and potent corticosteroid and potent corticosteroid vs a combined product containing vitamin D or vitamin D analogue and potent corticosteroid) that could potentially give inconsistent estimates of effect.

To overcome these problems, a hierarchical Bayesian network meta-analysis (NMA) was performed. This type of analysis allows for the synthesis of data from direct and indirect comparisons and allows for the ranking of different interventions in order of efficacy, defined as the achievement of clearance or near clearance. A network meta-analysis also provides estimates of effect (with 95% credible interval) for each intervention compared to one another and compared to a single baseline risk. These estimates provide a useful and coherent clinical summary of the results and facilitate the formation of recommendations based on the best available evidence. Furthermore, these estimates were used to parameterise treatment effectiveness of the topical therapies in the original cost-effectiveness modelling outlined in section 8.1.19. For details on the methods, results and interpretation of the network meta-analyses, see Appendix K.

The inclusion criteria for and intervention compared in the NMA were the same as in the review of direct evidence (section 8.1.1), except that the one study conducted entirely in children was included in the NMA only in a sensitivity analysis. A class effect was still assumed, but in order to reduce heterogeneity in the network of evidence, interventions were broken down by treatment frequency from the outset. In other words, once daily vitamin D or vitamin D analogue and twice daily vitamin D or vitamin D analogue were considered separate comparators in the NMA. Placebo/vehicle delivered once daily was also considered separately from twice daily placebo/vehicle.

The outcomes considered as part of the NMA were restricted to those measuring response:

  • Clear/nearly clear or marked improvement (at least 75% improvement) on Investigator’s assessment of overall global improvement (IAGI) or clear/nearly clear/minimal (not mild) on Physician’s Global Assessment (PGA)
  • Clear/nearly clear or marked improvement (at least 75% improvement) on Patient’s assessment of overall global improvement (PAGI) or clear/nearly clear/minimal (not mild) on Patient’s Global Assessment

Some included studies will have reported both outcomes, whereas some will have only included one or the other. For this reason, two networks of evidence were developed and analysed.

8.1.18.1. Results of NMA for investigator assessed outcome: clear/nearly clear (IAGI/PGA)

Thirty-four studies15,25,29,49,58,73,79,81,102,125,132,148,156,167,178,179,197,208,210,211,216,227,246,255,298,302,311,313,344,360,400,417,419,428 met the inclusion criteria for the base case network meta-analysis of the investigator assessed outcome of clear/nearly clear. Three further studies251,295,401 were included in a sensitivity analysis, the details and results of which can be found in Appendix K. Based on the GRADE quality ratings from the review of direct comparisons (section 8.1), the evidence included in the network meta-analysis ranges in quality from very low to moderate.

Figure 3 presents all the interventions included in the NMA as well as shows where there is direct evidence for a particular comparison and the number of studies that have included that comparison. For example, there are 7 studies reporting the outcome ‘clear’ or ‘nearly clear’ as measured by IAGI or PGA for the comparison of twice daily vehicle/placebo and twice daily vitamin D or vitamin D analogue. The diagram also highlights where there are gaps in the direct evidence. For example, there are no studies comparing a combined product containing vitamin D or vitamin D analogue and potent corticosteroid to concurrent vitamin D or vitamin D analogue and potent corticosteroid (one applied in the morning and one in the evening).

Figure 3. Clear or nearly clear – IAGI and PGA.

Figure 3

Clear or nearly clear – IAGI and PGA. Note: Solid lines indicate direct head-to-head comparisons and the colour indicates the number of trials per comparison included in the base case. Dashed lines indicate all head-to-head comparisons included (more...)

The results of the network meta-analysis in terms of the relative risk of each intervention compared to twice daily vehicle/placebo are presented in Table 62. It also gives a probability that the intervention is the most effective overall.

Table 62. Relative risks of clear/nearly clear on IAGI/PGA for all interventions compared to twice daily vehicle/placebo.

Table 62

Relative risks of clear/nearly clear on IAGI/PGA for all interventions compared to twice daily vehicle/placebo.

Evidence statements

Results of the network meta-analysis of randomised controlled trials indicate that, compared to twice daily vehicle/placebo, the following interventions are statistically significantly more effective at inducing clearance/near clearance as measured by the investigator or physician (IAGI/PGA):

  • Once and twice daily very potent corticosteroid
  • Once and twice daily potent corticosteroid
  • Once and twice daily vitamin D or vitamin D analogue
  • Once daily dithranol
  • Twice daily coal tar
  • Vitamin D or vitamin D analogue and potent corticosteroid (combined in one product)
  • Vitamin D or vitamin D analogue and potent corticosteroid (applied separately – one in the morning, one in the evening)

Results of the network meta-analysis of randomised controlled trials indicate that, compared to twice daily vehicle/placebo, the following interventions are not statistically significantly more effective at inducing clearance/near clearance as measured by the investigator or physician (IAGI/PGA):

  • Once daily retinoid
  • Once daily coal tar

Results of the network meta-analysis indicate that there are very few comparisons between active treatments (i.e. anything other than vehicle/placebo) for which the treatment effect reaches statistical significance. A few exceptions include:

  • Once daily product containing calcipotriol monohydrate and betamethasone dipropionate is more effective than once daily vitamin D or vitamin D analogue, once daily potent corticosteroid and once daily retinoid.
  • Twice daily very potent corticosteroid is more effective than once daily retinoid and once daily dithranol.
  • Twice daily vitamin D or vitamin D analogue, twice daily potent corticosteroids, twice daily very potent corticosteroids, combined and concurrent vitamin D or vitamin D analogue and potent corticosteroids are all more effective than once daily coal tar.

Results indicate that there is a non-statistically significant trend for twice daily application of any topical to be more effective than once daily application of the same topical.

Details of the pairwise comparisons from the network meta-analysis can be found in appendix K.

8.1.18.2. Results of NMA for patient assessed outcome: clear/nearly clear (PAGI)

Fourteen studies29,66,79,125,132,141,179,197,198,208,216,298,360,417 met the inclusion criteria for the base case network meta-analysis of the patient assessed outcome of clear/nearly clear. Two further studies295,302 were included in a sensitivity analysis, the details and results of which can be found in Appendix L. Based on the GRADE quality ratings from the review of direct comparisons (section 8.1), the evidence included in the network meta-analysis ranges in quality from very low to moderate.

Figure 4 presents all the interventions included in the NMA as well as shows where there is direct evidence for a particular comparison and the number of studies that have included that comparison. From the diagram, one can see that fewer studies have reported PAGI. There are 4 studies reporting the outcome of ‘clear’ or ‘nearly clear’ as measured by PAGI (in contrast to 7 studies reporting for IAGI or PGA) for the comparison of twice daily vehicle/placebo and twice daily vitamin D or vitamin D analogue.

Figure 4. Clear or nearly clear - PAGI.

Figure 4

Clear or nearly clear - PAGI. Note: Solid lines indicate direct head-to-head comparisons and the colour indicates the number of trials per comparison included in the base case. Dashed lines indicate all head-to-head comparisons included in the sensitivity (more...)

The results of the network meta-analysis in terms of the relative risk of each intervention compared to twice daily vehicle/placebo are presented in Table 63. It also gives a probability that the intervention is the most effective overall.

Table 63. Relative risks of clear/nearly clear with PAGI for all interventions compared to twice daily vehicle/placebo.

Table 63

Relative risks of clear/nearly clear with PAGI for all interventions compared to twice daily vehicle/placebo.

Evidence statements

Results of the network meta-analysis of randomised controlled trials indicate that, compared to twice daily vehicle/placebo, the following interventions are statistically significantly more effective at inducing clearance/near clearance as measured by the patient (PAGI):

  • Twice daily very potent corticosteroid
  • Once and twice daily potent corticosteroid
  • Twice daily vitamin D or vitamin D analogue
  • Vitamin D analogue and potent corticosteroid (combined in one product)
  • Vitamin D or vitamin D analogue and potent corticosteroid (applied separately – one in the morning, one in the evening)

Results of the network meta-analysis of randomised controlled trials indicate that, compared to twice daily vehicle/placebo, the following interventions trend toward being more effective at inducing clearance/near clearance as measured by the patient (IAGI/PGA), but the results fail to reach statistical significance:

  • Once daily vitamin D or vitamin D analogue
  • Once daily dithranol

Results of the network meta-analysis indicate that there are very few comparisons between active treatments (i.e. anything other than vehicle/placebo) for which the treatment effect reaches statistical significance. The one exception includes:

  • Once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate is more effective than once daily vitamin D or vitamin D analogue and more effective than once daily dithranol.

Details of the pairwise comparisons from the network meta-analysis can be found in appendix K.

8.1.19. Cost effectiveness evidence for trunk or limb psoriasis

8.1.19.1. Economic evidence – literature review

An economic evaluation should ideally compare all relevant alternatives. No applicable studies of good enough methodological quality were identified comparing all interventions of interest –vitamin D or vitamin D analogues, potent or very potent corticosteroids, coal tar, dithranol and retinoids – in the treatment of patients with mild to moderate chronic plaque psoriasis.

Three studies18,37,289 were identified that included two or more of the relevant comparators. These are summarised in the economic evidence profile below (Table 64 and Table 65). See also the full study evidence tables in Appendix I.

Table 64. Calcipotriol versus short contact dithranol – Economic study characteristics.

Table 64

Calcipotriol versus short contact dithranol – Economic study characteristics.

Table 65. Calcipotriol versus short contact dithranol – Economic summary of findings.

Table 65

Calcipotriol versus short contact dithranol – Economic summary of findings.

Six studies were selectively excluded, four due to very serious methodological limitations110,140,234,359 and two due to the availability of more applicable economic evidence21,310. Reasons for their exclusion are provided in Appendix G.

Ashcroft and colleagues present a simple decision tree analytic model to explore the relative cost effectiveness of topical calcipotriol and short contact dithranol. Caution should be exercised when interpreting the results of this study as it is unclear if the best possible sources were used to inform the parameters, and the short time horizon means that the costs of treatment failure may have not been fully accounted for.

Ashcroft et al. did not perform a quality of life assessment which limits its usefulness in determining cost effectiveness of the interventions studied. The below table shows the results of Ashcroft et al., with estimates of the possible incremental cost effectiveness ratio over a 1-year time horizon had quality of life measurements been incorporated. The ICERs presented below show that if utility gains of 0.03 or 0.09 are assumed (based on estimates used by other authors37,289 in the economic review) the additional cost of calcipotriol is very unlikely to be offset by the additional benefits associated with this treatment.

Table 66. Economic summary of Ashcroft et al. findings with quality of life incorporated.

Table 66

Economic summary of Ashcroft et al. findings with quality of life incorporated.

Both studies identified had potentially serious limitations with their chosen methodology. Bottomley and colleagues used an NHS provider perspective and was directly applicable, but is limited by the method used to generate estimates of treatment effect. The authors used performed an unadjusted indirect comparison which may introduce bias. The sensitivity analyses conducted by Bottomley et al. provide some indication that once daily product containing calcipotriol monohydrate and betamethasone dipropionate may be a cost effective strategy provided that the difference in utility between baseline and that experienced on the waiting list is small (i.e. 0.075). Interestingly, Bottomley and colleagues found concurrent but separate treatment with vitamin D or vitamin D analogue and potent corticosteroids to be the most expensive strategy and provided the least QALYs.

Table 67. Vitamin D or vitamin D analogues vs potent corticosteroids vs combined and concurrent vitamin D or vitamin D analogues and potent corticosteroids (one applied in the morning and one in the evening) - Economic summary of findings.

Table 67

Vitamin D or vitamin D analogues vs potent corticosteroids vs combined and concurrent vitamin D or vitamin D analogues and potent corticosteroids (one applied in the morning and one in the evening) - Economic summary of findings.

Oh and colleagues compared calcipotriol and different durations of clobetasol after first line treatment of a potent corticosteroid (betamethasone valerate) failed. Their evidence suggests that where the needed dosage and length of treatment of calcipotriol is similar or less than the ultra high potency corticosteroid clobetasol propionate, then calcipotriol might be the more cost effective second line treatment, however its incremental cost effectiveness compared to 2 weeks of very potent steroid was over the NICE £20,000 per QALY threshold. In a second analysis, they found that calcipotriol performed better as a second line treatment for psoriasis which had returned following prior treatment with betamethasone valerate or other agents, with increased utility due to lower side effects compared to fluocinonide.

8.1.19.2. Economic evidence – original economic analysis

The review of clinical evidence for topical therapies used in the treatment of individuals with mild to moderate plaque psoriasis showed that there were a wide variety of options – emollients, tars, dithranol, retinoids, corticosteroids (potent and very potent), vitamin D or vitamin D analogues and combination products – each associated with certain advantages and disadvantages. The results of the network meta-analysis suggested that some interventions, such as combined or concurrent vitamin D analogue and potent corticosteroid, were more likely to induce clearance or near clearance than others. Given that these combined and concurrent application strategies carry additional cost compared to both their individual constituent parts and compared to other topical alternatives, it was important to consider whether these additional costs are justified by additional health benefits in terms of improved quality of life.

The choice of which topical therapy to offer patients with mild to moderate psoriasis in primary care was identified as among the highest economic priorities by the GDG because the greatest proportion of psoriasis patients are managed at this point in the care pathway. Even if the unit costs of the interventions are quite modest, the population affected is relatively large; therefore the health economic impact of any recommendation is likely to be substantial.

Three cost-effectiveness analyses were identified in the published literature, but each had methodological limitations that called its conclusions into question. The analysis by Ashcroft and colleagues18 was based on only one trial and included only two of the interventions of interest (dithranol and calcipotriol). The analysis by Oh and colleagues289 was quite old and had analysed economic outcomes for different lines of treatment within separate models each having different comparators, thus making it difficult to identify a clearly cost-effective sequence of topical therapies. The analysis by Bottomley and colleagues,37 although the most applicable of the included studies, used an unadjusted indirect comparison to inform the treatment effect estimates, which likely overestimated the effectiveness of some interventions and underestimated the effectiveness of others. Bottomley and colleagues also did not include all the possible comparators of interest. Due to the methodological limitations of the published economic analyses, there was still substantial uncertainty as to which topical therapy or therapies represented the best value for NHS resources. In order to reduce this uncertainty, an original cost-effectiveness analysis was undertaken by the guideline health economist in collaboration with the GDG. Below is a summary of the analysis that was undertaken. For full details please see Appendix M: Cost-effectiveness analysis.

8.1.19.3. Methods

An analysis was undertaken to evaluate the relative cost-effectiveness of different topical therapy sequences used in the treatment of individuals with mild to moderate chronic plaque psoriasis. A Markov model was used to estimate 12-month costs and quality-adjusted life years (QALYs) from a current UK NHS and personal social services perspective. A 12-month time horizon was considered clinically relevant and sufficiently long enough to capture important costs and consequences of first-line treatment in primary care. Uncertainty was explored through probabilistic analysis and sensitivity analysis. The performance of alternative treatment sequences was estimated using incremental cost-effectiveness ratios (ICERs), defined as the added cost of a given strategy divided by its added benefit compared with the next most expensive strategy. A threshold of £20,000 per QALY gained was used to assess cost-effectiveness.

The aim of the analysis was to identify the most cost-effective sequence of first, second and third line topical therapies. It was important to model sequences given that most patients will commence treatment with one topical and then try others before moving on to more intensive treatments such as phototherapy and/or systemic therapy. In all, 118 sequences were compared in the base case analysis. Table 68 presents the list of possible first, second and third line treatments which may be combined in a sequence.

Table 68. All possible sequences of first, second and third line interventions.

Table 68

All possible sequences of first, second and third line interventions.

The following conditions were placed on the sequences, ensuring that they represented logical clinical practice:

  • Concurrent treatment with vitamin D or vitamin D analogue and potent corticosteroid (one applied in the morning and one in the evening) would not come after a failure of once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate;
  • Once daily treatment with a given topical would not come after a failure of twice daily treatment with the same topical;
  • Once daily treatment with potent steroid or vitamin D or vitamin D analogue would not come after concurrent treatment with vitamin D or vitamin D analogue and potent corticosteroid (one applied in the morning and one in the evening) or once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate;
  • No strategy could include potent corticosteroids among all three lines of treatment (including as part of concurrent vitamin D or vitamin D analogues and potent corticosteroid (one applied in the morning and one in the evening) or combined product containing calcipotriol monohydrate and betamethasone dipropionate).

Most comparators focus on evaluating a trial of three different treatments before referral for specialist review, but the GDG was also interested in whether earlier escalation of care might be more cost-effective. To test this, strategies have also been combined into two-treatment sequences with referral following a failure of second line treatment.

Due to the unacceptability of dithranol and coal tar as routine treatments (difficult application, risk of staining, strong and unpleasant odours, etc), these treatments were reserved for third line treatment only. This reflects their current placement in primary care given the availability of more acceptable and effective topicals such as those being compared as first and second line topicals. In a series of sensitivity analyses, other restrictions were placed on the potential sequences, namely due to concerns about the safety of continued use of potent corticosteroids.

The structure of the model developed by the NCGC was adapted from the model developed by Bottomley and colleagues37 and was validated by the GDG as a reasonable reflection of current clinical practice. The Markov model and how patients move through the pathway is illustrated in Figure 5. Key model assumptions (these are discussed in more detail in the full write-up in Appendix M):

Figure 5. Markov model of treatment with topical therapy.

Figure 5

Markov model of treatment with topical therapy.

  • All hypothetical patients commence treatment with a given topical and experience one of two outcomes after 4 or 8 weeks:
    • response (defined as clearance/near clearance of their psoriasis)
    • no response (defined as something less than clearance/near clearance of their psoriasis).
  • Patients who respond stop treatment and they either maintain response in the absence of treatment or they relapse.
    • Patients who relapse resume treatment with the same topical and again face a probability of responding or not responding.
  • Patients who do not respond to a given topical after 8 weeks of treatment are assumed to return to their GP and receive a prescription for an alternative topical therapy.
  • Patients can receive up to three different topical therapies before being referred by the GP to a specialist review in an outpatient dermatology clinic where second-line treatment options could be considered.
    • Some proportion of these referred patients will be kept on topical therapies, receive support and advice at the review consultation and be discharged back to their GP for long-term management.
    • The remaining proportion undergo a course of phototherapy:

      If they respond to phototherapy they are then discharged to their GP for long-term management.

      If they do not respond to phototherapy they continue to be managed by a specialist.

Movement between various health states is governed by transition probabilities, derived from the systematic review of clinical effectiveness data. Thirteen 4-week cycles were modelled, resulting in a 1-year time horizon for the analysis, with a half-cycle correction applied.

Model inputs were based on the clinical effectiveness review undertaken for the guideline, other published data and expert opinion where required. These are described in full in the technical report in Appendix M. All model inputs and assumptions were validated by the GDG.

8.1.19.4. Results

This analysis found that, given a NICE willingness-to-pay threshold of £20,000 per QALY gained, the most cost-effective strategy is likely to be one of starting with twice daily potent corticosteroid and moving to concurrent potent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening) and then twice daily coal tar. This strategy was also the least costly strategy among the 118 modelled. Base case results for non-dominated and non-extendedly dominated strategies are presented Table 69.

Table 69. Incremental analysis of base case results – psoriasis of trunk and limbs.

Table 69

Incremental analysis of base case results – psoriasis of trunk and limbs.

Results showed that starting with concurrent potent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening) and switching to twice daily potent corticosteroid and then twice daily coal tar is £9 more costly over 1 year and only produces 0.00041 more QALYs than the least costly strategy mentioned above. This gives it an incremental cost-effectiveness ratio (ICER) of £22,658 which is just above the NICE £20,000 per QALY threshold.

The most effective strategy (once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate then twice daily potent corticosteroid then twice daily coal tar) costs an additional £192 per year compared to the next most costly non-dominated strategy (concurrent steroid and vitamin D or vitamin D then twice daily potent steroid then twice daily coal tar), yet produces just 0.00107 additional QALYs for an ICER of over £179,000. Based on the results of this model, it appears that starting with once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate, although most effective, is very unlikely to be cost-effective.

Results of the analysis showed that a strategy of using vehicle or emollient with no active agent only was the most costly and least effective, largely driven by the cost of referrals and specialist management for non-responders. Strategies that included once or twice daily vitamin D or vitamin D analogue were not cost-effective regardless of where they were included in the sequence. This is largely due to their relatively low rank in terms of effectiveness and their relatively high acquisition cost. Strategies that included dithranol were also all dominated, that is more costly and less effective than alternatives. Finally, strategies in which patients were referred after non-response to only 2 topicals were all dominated, thus not cost effective.

The probabilistic analysis indicates that there is a great deal of uncertainty as to which sequence is optimal (i.e. most cost effective). There appears to be very little difference between initial potent corticosteroid followed by concurrent potent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening) and vice versa, with the difference in their net monetary benefits (NMB) being only £1 (£16,748 and £16,747 respectively) and both having an equal probability of being optimal at a £20,000 willingness to pay threshold. Generally, it looks as though a strategy of starting with either potent corticosteroids or concurrent treatment with potent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening) is most likely to be cost-effective, whereas starting with once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate is very unlikely to be cost-effective.

A series of sensitivity analyses suggested that the conclusions from the base case are sensitive to changes in some parameters and/or assumptions.

Sensitivity analyses – Treatment effects

The network meta-analysis of topical therapies was performed for two response outcomes: investigator assessed global improvement (IAGI) and patient assessed global improvement (PAGI). The economic evaluation used the investigator assessed outcome in the base case, largely because there was more data from the randomised evidence reported for this outcome. In a sensitivity analysis, treatment effects from the network meta-analysis of patient reported outcome was used.

Results of the analysis using patient reported outcomes indicates that starting treatment with once daily potent corticosteroids, moving on to the concurrent treatment if that fails and then trying twice daily vitamin D or vitamin D analogue is likely to be both the least costly and most cost-effective strategy given a threshold of £20,000 per QALY gained. Initial treatment with concurrent potent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening) appears less cost-effective using patient reported outcomes than physician reported outcomes, unlikely to be cost-effective at thresholds less than £100,000. Once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate, first or second line in a sequence, still looks to generate additional benefits (QALYs), but at additional costs unlikely to be considered good value for NHS resource (ICERs upwards of £115,000 per QALY gained).

The base case network meta-analysis of physician/investigator assessed response used in the base case cost-effectiveness analysis included all RCTs that met the inclusion criteria for the clinical review of direct evidence. The review of direct evidence was quite focused and as such did not include evidence for every possible pair wise comparison. In a sensitivity analysis of the network meta-analysis and thus the cost-effectiveness analysis, additional studies were included. For details on the particulars of these sensitivity analyses and what effect they had on the estimated treatment effects, see Appendix K.

When treatment effects were based on all relevant RCT data, the results of the base case changed only slightly. Twice daily potent corticosteroid followed by concurrent steroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening) is still likely to be optimal for first and second line treatments. However, instead of twice daily coal representing the optimal third line topical, twice daily vitamin D or vitamin D analogue looks to be most cost-effective. This sensitivity analysis calls into question whether vitamin D or vitamin D analogue or coal tar represents the better third line treatment option.

Sensitivity analysis – Variation in early versus late response

The base case assumed that patients would trial a given topical for up to 8 weeks and that some proportion of patients would be expected to respond by 4 weeks and discontinue treatment at that time. The remainder would carry on to 8 weeks, at which time non-responders would move on to the next topical in a sequence. The data defining the breakdown of early (at 4 weeks) vs late (at 8 weeks) responders was limited to two studies103,156 and GDG opinion and was thus very uncertain. Deterministic sensitivity analyses were performed around these parameters to observe the impact on the results.

First, an analysis was performed in which no one was expected to respond and discontinue treatment at 4 weeks (i.e. all responders require 8 weeks treatment). Compared to the results of the base case when all comparators are included, the rank order of strategies in terms of mean net benefits changed very little. The ICERs for strategies on the cost-effectiveness frontier (see Table 69) increased relative to the base case, thus becoming less likely to be considered cost-effective. This analysis found that, given a NICE willingness-to-pay threshold of £20,000 per QALY gained, the most cost-effective strategy is likely to be one of starting with twice daily potent corticosteroid and moving to concurrent potent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening) and then twice daily coal tar. This strategy was also the least costly strategy among the 118 modelled. Base case results for non-dominated and non-extendedly dominated strategies are presented Table 69.

Results showed that starting with concurrent potent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening) and switching to twice daily potent corticosteroid and then twice daily coal tar is £9 more costly over 1 year and only produces 0.00041 more QALYs than the least costly strategy mentioned above. This gives it an incremental cost-effectiveness ratio (ICER) of £22,658 which is just above the NICE £20,000 per QALY threshold.

The most effective strategy (once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate then twice daily potent corticosteroid then twice daily coal tar) costs an additional £192 per year compared to the next most costly non-dominated strategy (concurrent steroid and vitamin D or vitamin D then twice daily potent steroid then twice daily coal tar), yet produces just 0.00107 additional QALYs for an ICER of over £179,000. Based on the results of this model, it appears that starting with once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate, although most effective, is very unlikely to be cost-effective.

Second, an analysis was performed in which all responders were assumed to respond by 4 weeks, with no one requiring an additional 4 weeks of treatment. The ICER for all strategies on the cost-effectiveness plane (see Table 69) decreased relative to the base case, and now starting with concurrent therapy and moving to twice daily potent corticosteroids looks to be cost-effective at a £20,000 threshold compared to potent corticosteroids and then concurrent therapy. Initial treatment with once daily TCF product is still unlikely to be cost-effective, with an ICER of more than £140,000.

Finally, an analysis was performed in which a 4-week stopping rule was applied. In this scenario, responders were limited to those that have responded by week 4, and all other patients are assumed to move on to the next topical in the sequence (i.e. no one continues to 8 weeks of treatment with the same topical). Relative to the base case, the total costs for all strategies more than doubled as more patients were classified as non-responders and moved down the care pathway reaching referral to secondary care. Starting with concurrent therapy and then moving to twice daily potent corticosteroids was now the least costly strategy and most likely to be cost-effective. The ICER for once daily TCF product instead of concurrent therapy in this sequence decreased substantially relative to the base case (£174,000 to £94,000) but is still unlikely to be considered cost-effective at the NICE threshold.

Sensitivity analysis – Reduced adherence

There was some concern that issues of treatment adherence were inadequately captured in the model. The estimates of effect used in the base case were derived from randomised controlled trials which may represent the best case scenario for topical therapies. The GDG wished to explore how reduced adherence to twice daily treatments would affect the conclusions of the base case. In this scenario, 60% of patients being treated with twice daily topical were assumed to adhere to twice daily treatment whilst the remaining 40% of patients were assumed to apply the topical only once daily. For concurrent therapy, the 40% were assumed to adhere to once daily potent corticosteroid treatment only. Efficacy of the twice daily treatments would thus be reduced compared to the base case estimates. To be conservative, no reductions in cost were assumed despite the fact that less topical would be used.

With adherence reduced, there is no change substantive change to the results of the base case. Total costs across all strategies increase slightly (average of £27 more) and benefits decreased very slightly (average of 0.0007 fewer QALYs), but the conclusions from the base case remain unchanged. The most cost-effective strategy, given a £20,000 per additional QALY threshold is still twice daily potent corticosteroid followed by concurrent therapy and then twice daily coal tar. To put concurrent therapy before twice daily potent corticosteroids has an ICER of £36,000 (up from £23,000 in base case) and to replace concurrent therapy with once daily TCF before steroids has an ICER of £76,609 (down from £174,545 in the base case).

Sensitivity analysis – Utility values

In the base case, the mean utility gain associated with achieving some level of improvement, but not clearance or near clearance was assumed to be 0.05. This value was based on a downward adjustment of a value used in a recent cost-utility analysis included in the health economic review. Bottomley and colleagues37 modelled a utility gain of 0.07 for non-responders compared to baseline. To see what effect the GDG adjustment had on the results, the Bottomley figure (0.07) was used in a sensitivity analysis

Results indicate that the conclusion about cost-effectiveness changes very little using this more optimistic estimate of utility gain. The ICERs for all strategies increases relative to the base case; therefore, starting with concurrent treatment before twice daily potent corticosteroids is less likely to be cost-effective (ICER=£88,333 vs £23,250 in the base case). Similarly, the ICER for a strategy starting with combined product containing calcipotriol monohydrate and betamethasone dipropionate increased to over £787,000 compared to starting with concurrent treatment (£174,500 in the base case).

Sensitivity analysis – 4-week quantity of combined product containing calcipotriol monohydrate and betamethasone dipropionate

In the base case, hypothetical patients are assumed to use 134.0 g of combined product containing calcipotriol monohydrate and betamethasone dipropionate during 4 weeks of treatment. Bottomley and colleagues used a much lower value for this input (92.6 g), and we explored how the results of the NCGC analysis might change if this lower estimate was used. The cost of 92.6 g of combined product containing calcipotriol monohydrate and betamethasone dipropionate was £61.27 (compared to £94.26 in the base case). The results of this sensitivity analysis showed that the ICER for combined product containing calcipotriol monohydrate and betamethasone dipropionate improved compared to the base case (£124,400 vs £174,545); however this is still well above the NICE cost-effectiveness threshold of £20,000 per additional QALY. Initial therapy with twice daily potent corticosteroid or concurrent vitamin D or vitamin D analogue and potent corticosteroid (one applied in the morning and one in the evening) is still more likely to be considered cost-effective.

Sensitivity analysis – Unit cost of potent corticosteroids and vitamin D and vitamin D analogues

The base case assumed that the cost for each topical was based on the product and formulation with the lowest unit cost per gram/millilitre. Given that clinicians and patients may have preferences for different products or formulations, it was considered necessary to explore how variation in price of topicals, particularly potent corticosteroids and vitamin D, might affect the results. To do this, the highest cost (per gram) potent corticosteroid Synalar gel (fluocinolone acetonide) was assumed in place of Betnovate cream or ointment. The cost of Synalar gel is around four times that of Betnovate cream/ointment. In another analysis, the most costly vitamin D ointment, Curatoderm (tacalcitol), was assumed instead of Silkis (calcitriol). The cost of Curatoderm is around 2.5 times more costly than Silkis and 1.6 times more costly than Dovonex (calcipotriol) ointment. In a final sensitivity analysis, both Synalar gel and Curatoderm were used.

Sensitivity analyses – Restricted comparators

The base case analysis put a several conditions on the way topicals could be sequenced (see Table 68 in section 8.1.19.3). These conditions did not restrict how potent corticosteroids were fit into treatment sequences other than that they could not appear in all three lines of treatment. This included their use as part of concurrent or combined treatment. The GDG expressed concern that these restrictions may not fully reflect the caution they would use in prescribing trials of potent corticosteroids, in that the BNF discourages continuous use of potent corticosteroids for more than 8 weeks at a time. The GDG was also concerned that the analysis did not fully capture the safety risks associated with the continuous or intermittent use of twice daily potent steroids. In a series of sensitivity analyses, various additional restrictions were placed on the treatment sequences.

In the first scenario, it was assumed that interventions that included potent corticosteroids could not be offered consecutively. For example, once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate could not be offered after treatment with once or twice daily potent corticosteroids, nor could twice daily potent corticosteroid follow once daily potent corticosteroid. Under this assumption, starting with twice daily corticosteroid, then trying twice daily vitamin D or vitamin D analogue and then using both potent corticosteroid and vitamin D or vitamin D analogue concurrently (one applied in the morning and one in the evening) would represent the best value for NHS resources given a £20,000 per QALY threshold. Starting with concurrent treatment would only be cost-effective at thresholds of greater than £33,000 and combined product containing calcipotriol monohydrate and betamethasone dipropionate would only be cost-effective at thresholds over £202,000.

In the second scenario, it was assumed that twice daily corticosteroid could not be prescribed as a first or second line topical therapy, but consecutive use of potent corticosteroids was permitted. Under this scenario, the optimal strategy was to start with concurrent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening), then try twice daily vitamin D or vitamin D analogue alone and finally twice daily potent corticosteroid only. This had an ICER of £18,000 per QALY gained compared to once daily potent corticosteroid followed by concurrent treatment and then twice daily coal tar. Strategies including combined product containing calcipotriol monohydrate and betamethasone dipropionate either as second or first line were not cost-effective unless the threshold was over £110,000 and £446,000, respectively.

A third scenario combined the first and second scenarios, such that twice daily potent corticosteroid could not be prescribed as first or second line treatment and no sequences could include consecutive lines of potent steroid containing strategies. Under these conditions, the same sequence as in scenario 2 is most cost-effective (concurrent – vit D BD – PS BD). Combined product containing calcipotriol monohydrate and betamethasone dipropionate replaces twice daily steroid in that sequence only if the threshold willingness to pay is £134,000 and replaces concurrent treatment in the same sequence if the threshold is £202,000.

In a fourth and final scenario, twice daily potent corticosteroid was removed entirely and no potent steroid containing products could be prescribed consecutively. Under this assumption, the most cost-effective sequence was initial concurrent treatment followed by twice daily vitamin D or vitamin D analogue alone and then twice daily coal tar. Combined product containing calcipotriol monohydrate and betamethasone dipropionate replaces twice daily coal tar in that sequence at a threshold of over £47,000 and replaces concurrent treatment at a threshold of over £489,000.

Sensitivity analyses – downstream resource use and cost

Changes to the assumed probability of referral to secondary care and proportion offered phototherapy have no meaningful effect on the conclusions of the base case. The probability of referral to secondary care was varied downwards to 40% and upward to 80%. When referral occurred less often than in the base case, there was no change to the rank order of strategies, but the ICER for a strategy where combined product containing calcipotriol monohydrate and betamethasone dipropionate was used first instead of concurrent treatment increased to £200,000 per additional QALY. When referral occurred more often than in the base case, there was still no change in the rank order, but the ICER for combined product containing calcipotriol monohydrate and betamethasone dipropionate was slightly lower. If the probability of undergoing UVB phototherapy upon referral was higher than in the base case (50% vs 30%), then the ICER for combined product containing calcipotriol monohydrate and betamethasone dipropionate compared to concurrent treatment reduced slightly, but not enough to make it cost-effective. Finally, if instead of assuming patients are treated with UVB phototherapy, it is assumed they receive outpatient day care treatment with specialist supervised topical therapies, then the ICER for concurrent therapy before potent corticosteroids alone increases to over £30,000 per QALY and the ICER for initial combined product containing calcipotriol monohydrate and betamethasone dipropionate instead of concurrent therapy decreases to £155,000 per QALY.

If the time horizon is extended for 2 to 3 years and cumulatively more patients see a specialist and move on to UVB phototherapy, then initial treatment with concurrent vitamin D or vitamin D analogue and potent corticosteroids (one applied in the morning and one in the evening) becomes more cost-effective than starting with potent corticosteroids alone. When the time horizon is extended, TCF product becomes more cost-effective compared to concurrent treatment (ICER = £118,067 at 2 years; ICER = £90,710 at 3 years; ICER=£75,255 at 5 years; ICER=£73,541 at 10 years), but is still very unlikely to be considered cost effective given the NICE willingness to pay threshold of £20,000 per QALY gained. Visual inspection of the health state membership probabilities over a 10-year time horizon indicates that patients are no longer transitioning between health states after 8 years because they have all reached long-term management with a GP or specialist by this point. This suggests that the ICER for TCF product is unlikely to come down any further even if the model time horizon is extended beyond 10 years.

8.1.19.5. Interpretation and limitations

In assessing the relative cost-effectiveness of alternative topical therapies in patients with mild to moderate psoriasis limited evidence was available from the published economic literature. The evidence that was identified and included in the health economic review had potentially serious limitations and therefore the GDG considered it a priority to undertake original evaluation for the guideline in order to inform recommendations. This analysis showed that there were relatively small differences in terms of benefit between different topical sequences, but the differences in terms of cost were quite substantial. Based on the mean costs and benefits, the analysis suggests that initial treatment with potent corticosteroids followed by concurrent treatment with potent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening) and followed then by twice daily coal tar therapy is likely to represent the most cost-effective sequence for implementation in primary care. Uncertainties in the analysis were explored through sensitivity analysis which showed that in some scenarios

  • Once daily potent corticosteroid or concurrent treatment should come first in the sequence
  • Twice daily vitamin D or vitamin D analogue should come second or third in the sequence, after concurrent treatment
  • Combined product containing calcipotriol monohydrate and betamethasone dipropionate should be offered third in the sequence, after potent corticosteroids and concurrent treatment

Sequences starting with once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate were slightly more effective than the same sequence starting with concurrent potent corticosteroid and vitamin D or vitamin D analogue (one applied in the morning and one in the evening); however, the very modest additional benefit (0.0011) would only be considered potentially cost-effective if willingness to pay thresholds were between £ 100,000 and £ 500,000 per QALY gained.

The analysis has several limitations which were considered carefully by the GDG. Firstly, the analysis evaluates treatment sequences even though the available trial data compares single topicals head to head without sequencing. In order to apply the treatment effects within the sequencing model, we assumed that treatment effects were independent. That is, we assumed the effectiveness of combined product containing calcipotriol monohydrate and betamethasone dipropionate as a second or third line topical was equal to its effectiveness as a first line agent and that this was true regardless of other topicals it may follow. The GDG did not believe this to be a significant limitation given that the patients included in the overwhelming majority of RCTs were reported to have psoriasis for longer than 5 years, during which the can be assumed to have previously tried, succeeded and/or failed various topical treatments.

The analysis only captured the efficacy of topicals and did not capture the costs or consequences of adverse events. Although the RCT evidence on adverse events was sparse, the GDG is conscious of the risks associated with the long-term use of potent and very potent corticosteroids. They carefully considered whether the added effect in terms of clearance was worth the potential risks of adverse effects.

The model was also focused on the induction of disease clearance as opposed to the maintenance of clearance. Trials focusing on maintenance were limited in number and inadequately reported for use in the economic model. In particular, there was uncertainty as to how maintenance treatments were applied in the trials and therefore incorporating such evidence and assumptions into the model was considered too difficult and unlikely to be valid.

The model also takes a relatively short time horizon considering that psoriasis is a chronic, long term condition for which patients may undergo treatment for many years of their lives. Frequency and severity of relapse, selection for and speed of onward referral, methods of self-management and long-term safety are all issues inadequately addressed in the evidence base and therefore translate into limitations of the economic analysis.

The model estimated the health gain for each treatment by mapping the change in PASI score to the EQ-5D based on observational evidence. However, it has been noted that several important areas of health-related quality of life for people with psoriasis are not directly assessed by the EQ-5D questionnaire226. Therefore it is possible that the EQ-5D may lack content validity for these patients. Research is ongoing in this area. But we note that even using a £ 30,000 per QALY threshold rather than £ 20,000 would not change the conclusions of our analyses. Therefore only if the EQ-5D is under-estimating health gain of one treatment compared to another by a considerable extent, could this pose a serious limitation.

8.1.19.6. Comparison with published studies

The findings from the NCGC original economic analysis are quite different from the results of the most similar published study by Bottomley and colleagues37. Bottomley and colleagues found 8 weeks of once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate to dominate other modelled strategies including once and twice daily vitamin D or vitamin D analogue followed by potent corticosteroid, potent corticosteroid followed by vitamin D or vitamin D analogue and 8 weeks of concurrent treatment with vitamin D or vitamin D analogue and potent corticosteroid (one applied in the morning and one in the evening). Although the analysis appears to have been executed well, the estimates of effect and resource use had limitations which called the conclusions of the analysis into question.

The biggest differences in the results of the NCGC analysis presented here and the analysis undertaken by Bottomley has to do with the treatment effect sizes used. In their analysis, concurrent treatment was found to be very ineffective, with just 14.9% of patients responding with a PASI75 compared to the combined product containing calcipotriol monohydrate and betamethasone dipropionate to which 50.3% of patients responded (RR=3.38). The NCGC analysis showed a much small difference between these treatments, with 65.1% of patients responding to concurrent treatment and 70.7% responding to The combined product containing calcipotriol monohydrate and betamethasone dipropionate (RR=1.09).

In addition, the estimate they used for quantity of topical used per 4-week treatment period was 92.6 g, compared to the estimate used in the NCGC analysis 134.0 g. Based on these estimates of resource use, the NCGC analysis assumes 4 weeks of the combined product containing calcipotriol monohydrate and betamethasone dipropionate costs £ 29.26 more than Bottomley and colleagues did. Furthermore, the difference between the combined product containing calcipotriol monohydrate and betamethasone dipropionate and concurrent treatment is different between the analyses. The additional cost of the combined product containing calcipotriol monohydrate and betamethasone dipropionate was £ 36.91 in Bottomley and more than twice that, £ 76.34, in the NCGC analysis. We performed a sensitivity analysis in which we assumed the same quantity of the combined product containing calcipotriol monohydrate and betamethasone dipropionate used by Bottomley and colleagues (i.e. 92.6 g, £ 61.27). The ICER for the combined product containing calcipotriol monohydrate and betamethasone dipropionate improved compared to the base case (£ 124,400 vs £ 174,545), but was still well above the NICE cost-effectiveness threshold of £ 20,000 per additional QALY.

The one thing that Bottomley and colleagues were able to capture that the NCGC analysis was not had to do with the potential disutilities associated with adverse events; however these inputs were not reported, were not included in their base case and, their impact on the results were not reported in full. The authors simply state that the influence of AEs ‘had no impact on the results.’

8.1.19.7. Evidence statements

  • One partially applicable study with potentially serious limitations found that short-contact dithranol may be more cost-effective than calcipotriol.
  • One directly applicable study with potentially serious limitations found that a combined product containing calcipotriol monohydrate and betamethasone dipropionate administered once daily may be more cost effective than concurrent but separate treatment with vitamin D or vitamin D analogue and potent corticosteroids (one applied in the morning and one in the evening) and both vitamin D or vitamin D analogue alone (once daily and twice daily) and potent corticosteroids alone (once daily).
  • One partially applicable study with potentially serious limitations found that six weeks of vitamin D or vitamin D analogue offered after a trial of potent corticosteroids is likely to be cost effective compared to four or six weeks of very potent corticosteroids offered after a trial of potent corticosteroids; however, it is less likely to be cost effective compared to two weeks of very potent corticosteroids.
  • One partially applicable study with potentially serious limitations found that vitamin D or vitamin D analogue offered after failure of potent corticosteroid is likely to be cost effective compared to continued treatment with alternative potent corticosteroids.
  • New economic analysis from a current UK NHS and PSS perspective comparing 118 different sequences of topical therapies found twice daily potent corticosteroids or concurrent treatment (one in the morning and one in the evening) with potent corticosteroid and vitamin D or vitamin D analogue to be the most cost-effective options for the first and second line treatment of patients with mild to moderate chronic plaque psoriasis. This conclusion was robust to the majority of sensitivity analyses undertaken.
    • The base case and sensitivity analyses showed that the choice of third line treatment in a given sequence was highly uncertain. Depending upon the data used and assumptions made, third line treatment with twice daily coal tar, twice daily vitamin D or vitamin D analogue or once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate was likely to be most cost-effective.

8.1.20. Recommendations and link to evidence

Recommendations on topical therapyTopical therapy
The treatment pathway in this guideline begins with active topical therapies. The GDG acknowledged that the use of emollients in psoriasis was already widespread and hence the evidence review was limited to active topical therapies for psoriasis. Please refer to the BNF and cBNF for guidance on use of emollients.

General recommendations
25.

Offer people with psoriasis topical therapy as first-line treatment. Offer second- or third-line treatment options (phototherapy or systemic therapy) at the same time when topical therapy alone is unlikely to adequately control psoriasis, such as:

  • extensive disease (for example more than 10% of body surface area affected) or
  • at least ‘moderate’ on the static Physician’s Global Assessment or
  • where topical therapy is ineffective, such as nail disease.

See also recommendations 14; 60; 81; 100; 102; 104 and 106.

26.

Offer practical support and advice about the use and application of topical treatments. Advice should be provided by healthcare professionals who are trained and competent in the use of topical therapies. Support people to adhere to treatment in line with ‘Medicines adherence’ (NICE clinical guideline 76).

27.

When offering topical agents:

  • take into account patient preference, cosmetic acceptability, practical aspects of application and the site(s) and extent of psoriasis to be treated
  • discuss the variety of formulations available and, depending on the person’s preference, use:
    -

    cream, lotion or gel for widespread psoriasis

    -

    lotion, solution or gel for the scalp or hair-bearing areas

    -

    ointment to treat areas with thick adherent scale

  • be aware that topical treatment alone may not provide satisfactory disease control, especially in people with psoriasis that is extensive (for example more than 10% of body surface area affected) or at least ‘moderate’ on the static Physician’s Global Assessment.
28.

If a person of any age with psoriasis requiring topical therapy has a physical disability, or cognitive or visual impairment offer advice and practical support that take into account the person’s individual needs.

29.

Arrange a review appointment 4 weeks after starting a new topical treatment in adults, and 2 weeks after starting a new topical treatment in children, to:

  • evaluate tolerability, toxicity, and initial response to treatment (including measures of severity and impact described in recommendations 8, 11 and 12)
  • reinforce the importance of adherence when appropriate
  • reinforce the importance of a 4 week break between courses of potent/very potent corticosteroids (see recommendation 34).

If there is little or no improvement at this review, discuss the next treatment option with the person.

30.

Discuss with people whose psoriasis is responding to topical treatment (and their families or carers where appropriate):

  • the importance of continuing treatment until a satisfactory outcome is achieved (for example clear or nearly clear) or up to the recommended maximum treatment period for corticosteroids (see chapter 8)
  • that relapse occurs in most people after treatment is stopped
  • that after the initial treatment period topical treatments can be used when needed to maintain satisfactory disease control.
31.

Offer people with psoriasis a supply of their topical treatment to keep at home for the self-management of their condition.

32.

In people whose psoriasis has not responded satisfactorily to a topical treatment strategy, before changing to an alternative treatment:

  • discuss with the person whether they have any difficulties with application, cosmetic acceptability or tolerability and where relevant offer an alternative formulation
  • consider other possible reasons for non-adherence in line with ‘Medicines adherence’ (NICE clinical guideline 76).
How to use corticosteroids safelyzz
33.

Be aware that continuous use of potent or very potent corticosteroids may cause:

  • irreversible skin atrophy and striae
  • psoriasis to become unstable
  • systemic side effects when applied continuously to extensive psoriasis (for example more than 10% of body surface area affected).

Explain the risks of these side effects to people undergoing treatment (and their families or carers where appropriate) and discuss how to avoid them.

34.

Aim for a break of 4 weeks between courses of treatment with potent or very potent corticosteroids. Consider topical treatments that are not steroid-based (such as vitamin D or vitamin D analogues or coal tar) as needed to maintain psoriasis disease control during this period.

35.

When offering a corticosteroid for topical treatment select the potency and formulation based on the person’s need.

36.

Do not use very potent corticosteroids continuously at any site for longer than 4 weeks.

37.

Do not use potent corticosteroids continuously at any site for longer than 8 weeks.

38.

Do not use very potent corticosteroids in children and young people.

39.

Offer a review at least annually to adults with psoriasis who are using intermittent or short-term coursesaaa of a potent or very potent corticosteroid (either as monotherapy or in combined preparations) to assess for the presence of steroid atrophy and other adverse effects.

40.

Offer a review at least annually to children and young people with psoriasis who are using corticosteroids of any potency (either as monotherapy or in combined preparations) to assess for the presence of steroid atrophy and other adverse effects.

Recommendations on topical therapy for psoriasis of the trunk and limbTopical treatment of psoriasis affecting the trunk and limbs
41.

Offer a potent corticosteroid applied once daily plus vitamin D or a vitamin D analogue applied once daily (applied separately, one in the morning and the other in the evening) for up to 4 weeks as initial treatment for adults with trunk or limb psoriasis.

42.

If once-daily application of a potent corticosteroid plus once-daily application of vitamin D or a vitamin D analogue does not result in clearance, near clearance or satisfactory control of trunk or limb psoriasis in adults after a maximum of 8 weeksbbb, offer vitamin D or a vitamin D analogue alone applied twice daily.

43.

If twice-daily application of vitamin D or a vitamin D analogue does not result in clearance, near clearance or satisfactory control of trunk or limb psoriasis in adults after 8–12 weeksbbb, offer either:

  • a potent corticosteroid applied twice daily for up to 4 weeks or
  • a coal tar preparation applied once or twice daily.
44.

If a twice-daily potent corticosteroid or coal tar preparation cannot be used or a once-daily preparation would improve adherence in adults offer a combined product containing calcipotriol monohydrate and betamethasone dipropionate applied once daily for up to 4 weeks.

45.

Offer treatment with very potent corticosteroids in adults with trunk or limb psoriasis only:

  • in specialist settings under careful supervision
  • when other topical treatment strategies have failed
  • for a maximum period of 4 weeks.
46.

Consider short-contact dithranol for treatment-resistant psoriasis of the trunk or limbs and either:

  • give educational support for self-use or
  • ensure treatment is given in a specialist setting.
47.

For children and young people with trunk or limb psoriasis considerccc either:

  • calcipotriol applied once daily (only for those over 6 years of age) or
  • a potent corticosteroid applied once daily (only for those over 1 year of age).
Future research recommendations12. In people of all ages with psoriasis:
  • How should topical therapies be used to maintain disease control i) safely; ii) effectively and iii) what are the health economic implications?
  • What are the risks of ‘real life’ long term corticosteroid use, are there particular people at risk and what strategies can be used to modify or avoid risks?
Relative values of different outcomesThe GDG considered the following outcomes:
  • clear/nearly clear (defined as at least 75% improvement, very mild or clear on a static scale)
  • duration of remission (relapse rate and time to remission)
  • withdrawal due to toxicity
  • withdrawal due to lack of efficacy
  • skin atrophy (reporting of skin atrophy was not by quantifiable methods).
The GDG prioritised the following outcomes for decision making:
  • clear/nearly clear (investigator and patient assessed)
  • duration of remission
  • withdrawal due to toxicity.
Based on the clinical and cost-effectiveness evidence, the GDG recommended potent corticosteroids applied once daily plus vitamin D or a vitamin D analogue applied once daily (applied separately, one agent applied in the morning and the other in the evening) as the first topical intervention. This was the most cost-effective and clinically sensible option based on the investigator and patient assessment of achieving clear or nearly clear status after very potent steroids had been excluded owing to safety concerns. There was no clinically significant difference between most interventions in terms of withdrawal due to toxicity as the absolute numbers were low and clear evidence regarding duration of remission was lacking.
Trade off between clinical benefits and harms
  • The superior efficacy of potent corticosteroids compared with vitamin D analogues might be outweighed by the risk of local side effects (e.g. irreversible skin atrophy), shorter duration of remission, destabilisation of psoriasis, and, although rare, the potential for systemic side effects of corticosteroid in those with extensive disease. It was also recognised that such risks might be compounded by repeat prescriptions being issued without assessment.
  • The GDG discussed the risks and benefits of corticosteroids and considered that given their marked efficacy and cosmetic acceptability, potent corticosteroids could be recommended for the treatment of chronic plaque psoriasis in primary care in the context of appropriate review and patient education. However, very potent corticosteroids could not be recommended due to concerns about the rebound effect, irreversible skin atrophy, the risk of repeat prescriptions being issued without monitoring and lack of long-term safety data. Based on the duration of the trials (mostly up to 8 weeks for potent and 4 weeks for very potent corticosteroids) and in line with the clinical experience of the GDG, it was agreed that to ensure safe use, potent corticosteroids should not be used continuously for more than 8 weeks and very potent corticosteroids for more than 4 weeks. The data showed that the levels of skin atrophy at this point did not demonstrate clinically relevant harm and the treatment response was beginning to plateau.
  • The majority of those who are likely to respond within 8 weeks to potent or very potent steroids would have done so by 4 weeks. Therefore, the initial treatment period recommended for these topical agents is 4 weeks.
  • A break of 4 weeks between courses of potent or very potent corticosteroids was recommended, as the GDG were aware of data suggesting that 3–4 weeks is required for skin regeneration. Non-steroid based topical treatments are recommended to maintain disease control during this break, as there was evidence to suggest that vitamin D analogues could maintain response following short-term treatment with a combined product containing potent corticosteroid and vitamin D analogue.
  • The GDG considered that appropriate assessment and review of efficacy and safety was critical. An early review to identify tolerability/side-effects and to identify complete non-responders is needed. Since the most rapid improvement is seen within the first 4 weeks, a review after 4 weeks was agreed. Those with little or no improvement at 4 weeks should be cycled on to the next stage of the topical treatment pathway.
  • Although the combined product is not cost-effective for the average patient, it was considered an important third-line topical option. This was because in non-responders to topical treatment, concordance is often a problem and a once-daily, well-tolerated topical preparation would be of benefit in achieving clearance, so avoiding hospital referral and saving cost in this small group. Therefore, the GDG agreed to recommend once daily combined potent corticosteroid and vitamin D analogue in patients in whom twice daily potent corticosteroid or coal tar cannot be used and a once daily preparation would improve adherence.
Economic considerationsThe GDG relied on a variety of sources in their consideration of the costs and benefits of alternative topical therapies in the treatment of patients with mild to moderate psoriasis. Limited evidence, both in terms of quantity and quality, was identified in the published literature. The available evidence showed:
  • short-contact dithranol likely to be more cost-effective than a vitamin D analogue 18;
  • vitamin D analogue to be more cost-effective than potent and very potent corticosteroids 289; and
  • two compound formulation steroid and vitamin D analogue to be more cost-effective than concurrent (morning/evening) application of the two topicals and more cost-effective than both potent steroids and vitamin D analogues applied alone37
However, the GDG remained uncertain about the robustness of these conclusions.

Original decision modelling was undertaken for the guideline and showed that there were relatively small differences in terms of benefit between different topical sequences, but large differences in terms of cost. Based on the mean costs and benefits of 118 compared sequences, the analysis suggests that treatment with potent corticosteroids or concurrent treatment with potent corticosteroid and vitamin D analogue (morning/evening application) was likely to be the most cost-effective option for the first and second-line treatment of patients with mild to moderate psoriasis.

The analysis specifically found twice daily potent corticosteroid to be highly cost-effective, but the GDG expressed concern that the well known side effects of potent corticosteroids (e.g. skin atrophy, rapid relapse) were not adequately captured in the economic model owing to a lack of data. Twice-daily potent corticosteroids were more cost-effective than once-daily, largely because the quantities of topical used for once and twice daily application were very similar, yet the network meta-analysis showed a non-significant trend toward twice daily being more effective in the investigator assessed outcomes used in the base case scenario (OR=1.807, 95% CrI 0.42 to 8.074). However, this trend was reversed for the patient-assessed outcome, i.e. twice-daily performed less well than once-daily (OR=0.714, 95% CrI 0.14 to 3.549). This finding is reflected in the results of the sensitivity analysis where patient-reported response was used, which show once-daily to be more cost-effective than twice-daily. The consensus of the GDG was that they were uncertain that twice-daily potent corticosteroids were more effective than once-daily potent corticosteroids. They concluded that even if twice daily application was more effective at inducing clearance or near clearance than once daily application, the risks of higher dose steroids were very likely to outweigh the potential benefits and make the intervention less cost-effective. The GDG recommend that in the choice of potent corticosteroid formulation, consideration be given to patient preference, cosmetic acceptability, practical aspects of application and the site(s) and extent of psoriasis to be treated. The lowest cost preparation might not be the most cost-effective one if adherence is low.

Concurrent treatment with potent corticosteroid and vitamin D analogue (morning/evening application) was also likely to be cost-effective in a range of scenarios. In some cases, it was found to be a cost-effective first line treatment; however, the GDG felt this was too aggressive a strategy to start with for the majority of people with mild to moderate psoriasis being seen in primary care. Based on that, they concluded that the addition of once daily vitamin D analogue to once daily application of potent corticosteroid should be the next treatment offered if a potent corticosteroid alone has failed to induce the desired level of response. The GDG specifically considered whether they should offer concurrent treatment (morning/evening) with two separate topicals or offer combined treatment in a single product for use just once daily. They considered the results of the cost-effectiveness analysis which showed that combined treatment (once daily TCF product) is not cost-effective compared with concurrent treatment. This is because the network meta-analysis found them to have similar efficacy, but TCF product is much more costly (unit cost of 120g combined product containing calcipotriol monohydrate and betamethasone dipropionate is between 2 and 4 times more costly than the combined unit cost of 100g of vitamin D analogue and potent corticosteroid each). This is true even when the most costly potent corticosteroid and vitamin D products and formulations are assumed to be prescribed. The GDG considered whether a once daily application of the combined product may be cost-effective when considering the problems many patients have adhering to twice daily treatment regimens. The results of a sensitivity analysis wherein 40% of patients prescribed concurrent therapy were assumed to apply only their potent corticosteroid once per day showed that the very small benefits of once daily combined product were still outweighed by its extra cost. The GDG concluded that the combined formulation product as first-line treatment produced enough additional benefit to justify its substantial additional cost.

The base case cost-effectiveness analysis and sensitivity analyses showed that the choice of third line treatment in a given sequence was highly uncertain. Depending upon the data used and assumptions made, third line treatment with twice daily coal tar, twice daily vitamin D analogue or once daily TCF product was likely to be most cost effective. To reflect the uncertainties in the conclusions about cost-effectiveness and provide prescribers and patients with a degree of choice, the GDG chose to recommend all of these interventions if the patient has failed to achieve clearance or near clearance with potent corticosteroids alone or concurrent treatment with potent steroids and vitamin D analogue. They considered that some people may not choose to use coal tar as it has a pungent odour and that some people may prefer vitamin D analogues as they are generally safe for long term use. They considered that the combined potent corticosteroid and vitamin D analogue product was much more costly than other alternatives, but it may represent value for NHS resource in a select group of patients with resistant mild to moderate psoriasis. It also may be more cost-effective to offer if the alternative is referral and escalation of treatment to much costlier interventions (e.g. phototherapy, specialist applied topicals, systemic therapy, biologic therapy).

The cost-effectiveness analysis did not find short contact dithranol to be more cost-effective than other first, second and third line alternatives in the base case or any sensitivity analyses. The GDG did not want to rule dithranol out as a treatment option for some patients, but considered it only potentially cost-effective for patients who have failed to respond to other more efficacious and easy-to-use topical therapies. They emphasised the need for the healthcare professional to clearly explain proper application of dithranol for home use in order to maximise its effectiveness and reduce the inconvenience. They also considered that dithranol may be best delivered as part of treatment in a day care setting with specialist nurse supervision.

The cost-effectiveness of very potent corticosteroids was not evaluated as part of the decision-modelling, as the GDG did not consider it a safe treatment option for the management of mild to moderate psoriasis in primary care. They considered that based on its efficacy and relatively low cost (100g cream or ointment = £ 7.90), it was likely to represent good value for NHS resource so long as it is used with caution and under careful supervision of a specialist in secondary care.

In thinking about the potential risks of prescribing potent, and in select cases very potent corticosteroids, the GDG considered it essential to build in monitoring to assess efficacy and adverse events. The time horizon of the economic model was too short (1 year) to explicitly consider annual monitoring in the long term; however, it is very likely that the extra cost of an annual GP or specialist visit would be offset by the avoidance of irreversible adverse events that are associated with inappropriate and unsafe use of corticosteroids.

The cost-effectiveness of topical treatments for children was not explicitly considered in the decision modelling undertaken for the guideline; however, the GDG considered the results broadly applicable to this population. They considered that once-daily applications in children were likely to be more appropriate, and that evidence of effectiveness for combination strategies were lacking. Therefore, they concluded that for children with mild to moderate psoriasis, once daily application of potent corticosteroids or vitamin D analogue were likely to represent the best value for NHS resource. They also considered how infrequently psoriasis occurs in children and that referral to secondary care may be justified.
Quality of evidenceThe GDG noted variations in methodology and reporting between the studies, in particular:
  • frequency of administration of treatment
  • duration of follow-up
  • within (left-and right-hand side comparison) and between patient randomisation
  • topical formulation
  • baseline disease severity -of the studies that reported disease severity at baseline, 16 studies included moderate to severe disease. This does not reflect clinical practice as monotherapy with topicals is usually used to treat mild/moderate disease, which was the population of interest for this question.
Note that within-and between-patient studies have been pooled together in the analysis, and none of the studies reported sufficient information to take account of the within-patient correlation in the analysis. It was often not possible to say if consistent differences were present as there was only one within patient study in the comparison. When it was possible to assess this, no consistent difference was seen for efficacy outcomes, although for vitamin D analogues vs. placebo there may be a difference for between-and within-patient studies for withdrawals due to adverse events or lack of efficacy. For withdrawals due to adverse events, 5/6 between patient studies favoured vitamin D analogues (RR = 0.54) compared with 5/5 within patient studies210,211,311,354,411 which favoured placebo (RR = 3.00). Conversely, for withdrawals due to lack of efficacy 3/3 between patient studies favoured vitamin D analogues (RR = 0.15) whereas 3/3 within patient studies showed no difference (RR=1.00). However, the absolute number of withdrawals was low so this difference is unlikely to be clinically meaningful.
Vitamin D analogues vs placebo
The GDG noted that:
  • the Perez study gave an outlying result for the outcome of investigator’s assessment of global improvement (IAGI), and that there was a zero success rate in the placebo arm. This was considered unusual as emollients tend to have some level of efficacy.
  • the Langner 1993 study used an unlicensed dose of calcitriol (15μg twice daily).
Vitamin D analogue vs. corticosteroid
  • There was heterogeneity between the studies for the outcome of investigator’s assessment of improvement. This could not be explained by excluding studies at higher risk of bias or by any of the pre-defined subgroups for investigation, as a statistically significant level of inconsistency still remained. However, it appeared that betamethasone valerate was less effective than betamethasone dipropionate when compared with vitamin D analogues.
  • The GDG noted that the rates of remission were low for all interventions in the Fleming 2010A study but no clinical or methodological explanation could be found for this.
Vitamin D analogues vs. coal tar
  • There was significant heterogeneity between the studies. The heterogeneity may be explained by variation in treatment duration and coal tar taking longer to act than vitamin D analogue, so becoming relatively more effective at later timepoints. One of the studies (Pinheiro 1997) used a tar combination that includes a mild potency corticosteroid (alcoholic coal tar extract 5%, hydrocortisone 0.5%, allantoin 2%).
Maintenance therapy
Just two studies178,195,196 directly assessed maintenance treatment:
  • The Katz study had a maximum treatment period of 6 months with potent corticosteroid or placebo (using an intermittent regimen of 3 applications 12 hours apart once a week) among those who had achieved remission after 3–4 weeks treatment with a potent corticosteroid. The GRADE ratings for this study were low to moderate, and the definition of response was broader than that specified in the review protocol, which may over estimate efficacy (clear/slight improvement on a four point scale) but was included given the paucity of maintenance studies.
  • The Kragballe study had a 52 week treatment period for as-needed application of either a combined potent corticosteroid and vitamin D analogue, the combination for 4 weeks then calcipotriol for 48 weeks, or alternating 4 week periods of the combination product and calcipotriol. The one year timeframe of this study reflects clinical practice; however, the study was primarily designed to investigate safety rather than efficacy.
Treatment regimens
There were also 3 studies (Kragballe 2004, Ortonne 2004 and Saraceno) that assessed different treatment schedules (e.g. combination of potent corticosteroid and vitamin D analogue then vitamin D analogue alone) but these were only 16–24 weeks in duration and therefore of limited relevance.
The GDG noted that there was inconsistency between the studies for time to relapse and relapse rates during a post-treatment withdrawal phase among those who had achieved remission, and that only 4 studies reported these data (Langley 2011A, Camarasa 2003, Alora-Palli 2010, and Christensen 1999) and one during a maintenance treatment phase following remission (Katz 1991). Additionally, in the placebo group the numbers who achieved remission and were followed-up were very few and they may have gone into spontaneous remission; therefore, the time to relapse in this group may be a spurious result. As such, the GDG gave little weight to these data. The GDG noted that, in accordance with clinical experience, relapse rates following use of vitamin D analogues appeared to be lower than that with potent corticosteroids (although the time to relapse was similar in both groups).

Time to maximum effect
  • The GDG noted the following variables between the studies investigating time to remission/maximum effect which made interpretation and synthesis of the results difficult:
    • drug dosing
    • formulation
    • treatment duration
    • outcome measure.
  • The GDG also noted that the majority of the trials were not long enough to see the maximum effect. The only longer term study (Perez) was a 12 month follow up after randomised phase of trial. However, it included small numbers of participants (22 at the start with 6 remaining at the end) and so was excluded from the review.
  • The following maximum responses were noted:
    • for vitamin D or vitamin D analogues -was seen at 8–12 weeks (most rapid improvement was seen over the first 4 weeks).
    • for potent steroids -was not seen during the 8 week study period although continued improvement was likely to be minimal (most rapid improvement was seen over the first 2–4 weeks).
    • for very potent steroids -was not seen by end of 4-week study period although continued improvement was likely to be minimal (most rapid effect is seen over the first 2 weeks).
    • for the combined product -was at 12 weeks although the majority of this occurred within the first 4 weeks.
    • it was not possible to be sure about when the maximum response to coal tar preparations is seen owing to the different results between preparations and the paucity of evidence so no time frame for use is stated.
  • All treatment modalities demonstrated some efficacy by four weeks. The GDG agreed that based on the times to response, assessment at four weeks would be helpful to assess treatment efficacy, potential problems with use such as formulation, tolerability, cosmetic acceptability and to plan ongoing treatment strategy including treatment switch in the event of an inadequate response.
Relapse
From the evidence, relapse occurs in 20–80% of people following treatment withdrawal regardless of the specific topical treatment used, so the GDG agreed there should be an over-arching recommendation about offering strategies that recognise that repeat treatment is likely to be required and that patients need education on what to expect from treatment. Limited data on maintenance strategies precluded making separate recommendations on induction and maintenance of remission. In the absence of evidence, topical therapies should be continued to be prescribed and used ‘as needed’.
Treatment frequency
In considering differences between once and twice daily applications of potent corticosteroids, whilst there is generally a trend towards better efficacy with twice daily application, greater numbers of withdrawals due to adverse events were seen with twice daily potent corticosteroid compared with once daily. Therefore the GDG agreed that in view of convenience to the patient, potential cost benefit, and reduced risk of side effects especially in relation to corticosteroid use, once daily applications should be recommended in the first instance. Treatment could be escalated to twice daily if once daily is not effective.
Evidence gaps
The GDG noted important gaps in the evidence required to inform clinical practice. Psoriasis is a long term condition, but the vast majority of studies are 12 weeks or shorter in duration. Only limited data were available on longer term use, especially regarding the safety of very potent and potent steroids, treatment strategies for maintenance of disease control, relative benefits of the different interventions with respect to relapse and remission rates, and the value of early intervention (for example use of a topical treatment at first signs of disease occurrence).
Other considerationsGroups for special consideration
  • There are no groups of people who should not be offered topical therapy.
  • For patients with severe chronic plaque psoriasis (i.e. BSA>10% and/or PASI >10), self-administered topical treatment alone is unlikely to provide adequate disease control, is difficult from a practical point of view, and application of potent corticosteroid over large areas of inflamed skin may increase the risk of both local and systemic side effects. It was therefore agreed that additional treatment strategies should be routinely offered to this group.
  • Psoriasis is not common in children and therefore quicker escalation to secondary care may be appropriate. Giving GPs the option of using emollients and then referring without trying any active treatments was felt to be limiting. Plaques are usually thinner and less scaly in children. Topical calcipotriol is licensed for children above 6 years old. One study investigating calcipotriol in children found a smaller response compared to the adult studies, although this was one study with small numbers. From GDG experience, mild to moderate potency corticosteroids are also useful in children with or without tar but there was no evidence for this. Taking into consideration all of the above points, it was agreed that children should be reviewed after two weeks, as the plaques tend to be thinner and treatments should only be used within the licensed indications for trunk and limb psoriasis in children. For more information on dosing, in the absence of evidence, prescribers should refer to the BNF for Children.
  • The GDG discussed the needs of older people, people with limited mobility and people with psoriatic arthritis. It was noted that specialist help with application can improve outcomes for these groups of people.
Adherence
  • The GDG considered factors that may impact on treatment adherence and outcomes including cosmetic acceptability and local side effects.
  • For pragmatic reasons, the GDG had agreed that data on the impact of formulation on treatment outcomes would not be considered. However, it was agreed that formulation should always be considered when prescribing topical therapy to optimise treatment adherence and minimise local adverse effects. For example, a light cream or lotion may be appropriate for widespread, multiple small plaques to cover requisite large areas, lotions/solutions for hair bearing areas and ointments for scaly areas. It was noted that knowledge in primary care may be limited in this regard and simple guidance would be helpful. The GDG agreed that a specific recommendation about the need to consider formulation and cosmetic acceptability when prescribing topical therapy was justified.
  • Non-concordance should be considered if there is no response to treatment in line with ‘Medicines adherence’ (NICE clinical guideline 76)265
Safety and toxicity
  • There is enduring concern amongst clinicians and patients about potential risks of corticosteroids for the treatment of psoriasis including local skin atrophy, rapid relapse/rebound on treatment cessation, destabilisation of disease (for example induction of pustular psoriasis) and potentially systemic side effects in people with very widespread psoriasis, especially given that for chronic plaque psoriasis at most body sites (excluding face, flexures) potent or very potent corticosteroids are required to achieve clearance.
  • From GDG knowledge, vitamin D analogues, tar and dithranol do not cause skin atrophy whereas corticosteroids do.
  • The majority of the data on withdrawals and skin atrophy across all comparisons showed low event rates that gave very imprecise relative estimates, but in absolute terms demonstrated precise evidence of no clinically relevant difference between the interventions because the numbers involved were so low
  • Overall, the evidence did not indicate any statistically significant increased risk of steroid atrophy with corticosteroid use (potent and very potent) and the numbers of cases of skin atrophy reported were very low. The majority of cases of skin atrophy that were reported were in patients who received corticosteroids. However, this outcome was not reported in all studies and no study reported having used a reliable quantitative measure to assess the level of atrophy. It was noted that the lack of a significantly increased risk may be due to lack of appropriately designed studies of sufficient duration and power rather than lack of risk.
  • The GDG discussed whether extrapolation from the amount of steroid likely to be used if the Finlay fingertip unit was adhered to (i.e. 0.5g covers the equivalent of 2% BSA (or 2 ‘hands worth’)) could inform the likely safety of the recommendations made. Based on the fact that potent corticosteroid would only be the main treatment option in people with localised disease, with secondary care escalation immediately if BSA is >10%, a conservative assumption would be that anyone with a BSA of <10% may be managed with corticosteroids alone. Therefore, once daily application to 10% BSA (based on the fingertip unit) is equivalent to 2.5g daily application (75g per month). The GDG agreed that this is in keeping with dermatology practice that >100g month indicates potentially dangerous amounts of steroid and is less than the volumes used in the RCTs reviewed, which had few cases of atrophy. Therefore, the GDG were satisfied that based on the available evidence and current expert opinion the recommendations should not cause an increase in steroid-related toxicity in people with psoriasis.
  • Some patients may prefer to use topical therapies that do not contain corticosteroids (tar, dithranol, vitamin D analogues) due to concerns about corticosteroid side effects.
  • Tazarotene may be unpleasant to use. It causes burning and irritation of the skin (which was indicated by the evidence for a statistically significantly higher number of withdrawals due to adverse events among those treated with tazarotene compared with placebo in 2 studies), and shows only limited efficacy (approximately 6% achieved clearance or near clearance).
  • Dithranol is difficult to use at home due to staining, risk of burning unaffected skin and difficulties with self application, but is useful for large, thick, treatment resistant plaques. Educational support is required when prescribed.
Current practice
  • In primary care topical vitamin D analogues are considered the standard treatment. Combined potent corticosteroid/vitamin D analogue preparation is not in most GP formularies due to the cost. Most patients benefit from an emollient to relieve pruritus and scaliness.
  • PASI and DLQI are not used in primary care so could not be recommended for assessment of response to treatment. In addition sensitivity to change with PASI is poor in mild to moderate disease.
Other considerations from the evidence
  • The evidence suggested that time to relapse was shorter with potent and very potent corticosteroids compared to vitamin D analogues, tar and dithranol.
  • The GDG noted that in studies that compared various treatment sequences (e.g. combined product containing calcipotriol monohydrate and betamethasone dipropionate followed by either vitamin D alone or alternating vitamin D alone and combined product containing calcipotriol monohydrate and betamethasone dipropionate) with vitamin D alone for the full trial period if a combined product containing calcipotriol monohydrate and betamethasone dipropionate was present anywhere in the sequence, even just for the first 4 weeks, the efficacy was improved compared with vitamin D alone. The data suggested that this increased efficacy could be maintained by subsequent use of vitamin D analogue alone.
zz

See recommendations 56 and 58 for details on safe use of steroids at facial, flexural and genital sites.

aaa

See recommendations 36 and 37 for details on safe duration of steroid use.

bbb

See recommendation 32 for additional considerations before changing to the next treatment option.

ccc

Please refer to the BNF for children for information on appropriate dosing and duration of treatment.

8.2. Topical therapies for high impact or difficult sites

8.2.1. Methodological introduction

A literature search was conducted for RCTs or systematic reviews that compared the efficacy and safety of topical vitamin D and vitamin D analogues, mild to very potent corticosteroids, combined vitamin D or vitamin D analogue and potent corticosteroid or concurrent vitamin D or vitamin D analogue and potent corticosteroid (one applied in the morning and one in the evening), pimecrolimus, tacrolimus, tar, dithranol and retinoids in people with psoriasis at high impact and difficult to treat sites for the induction or maintenance of remission. The sites included were scalp, face and flexures (including genitals), which would be considered separately if stratified data were available.

No time limit was placed on the literature search and there were no limitations on duration of follow-up. However, indirect populations were excluded and the sample size had to be at least 25 participants in each arm.

The comparisons considered were any of the topical therapies compared with each other or with placebo/vehicle, while studies only comparing different dosages or formulations of the same intervention were excluded. Similarly, studies comparing interventions within the classes of either vitamin D or vitamin D analogues or corticosteroids were excluded (unless the comparison is for frequency of administration e.g., once or twice daily dosing). This is because we assume a class effect for these agents and so data on all vitamin D or vitamin D analogues was pooled into one analysis as was data on any potent corticosteroids and on very potent corticosteroids, unless heterogeneity was found.

The outcomes considered were:

  • Clear/nearly clear or marked improvement (at least 75% improvement) on Investigator’s assessment of overall global improvement (IAGI) or clear/nearly clear/minimal (not mild) on Physician’s Global Assessment (PGA)
  • Clear/nearly clear or marked improvement (at least 75% improvement) on Patient’s assessment of overall global improvement (PAGI) or clear/nearly clear/minimal (not mild) on Patient’s Global Assessment
  • Percentage change in PASI
  • Change in DLQI
  • Duration of remission
  • Time-to-remission or time-to-maximum effect based on IAGI, PGA or total severity score (to address part ii of the question)*
  • Withdrawal due to toxicity
  • Withdrawal due to lack of efficacy
  • Skin atrophy

Time-to-remission or time-to-maximum effect, absolute time-to-effect data or data from multiple time points in one study were reported as the first preference. Graphical data were only reported for interventions where such data were unavailable, or for long-term data not otherwise available. Additionally, data on IAGI, PGA or PAGI were reported in preference to TSS where available.

Twenty one RCTs48,108,109,128130,166,168,169,189,199,215,220,228,245,292,315,330,377,405,408 were found that addressed the question and were included in the review:

  • 18 of these studies48,108,109,128,130,166,168,169,189,199,228,245,292,315,330,377,405,408 addressed scalp psoriasis
  • One study129 addressed flexural psoriasis alone
  • Two studies215,220 addressed both face and flexural psoriasis
  • Two studies228;315 assessed long-term/maintenance treatment
  • No studies were available to address the use of topical treatments at high-impact or difficult to treat sites in children

A published Cochrane Review238 was available but was in the process of being updated by the Cochrane Review Group (and anticipated publication was outside of the development period of this guideline). The NCGC was unable to update the original Cochrane Review owing to differences in the outcomes required to feed in to a novel NCGC health economics model. The Cochrane review was used for NCGC cross referencing purposes and close collaboration between the Cochrane Review Group and NCGC meant that literature search strategies/protocols were shared. The Cochrane literature search was re-run and updated to include papers to the present day. Additionally, it was possible to use some of the data extracted on study characteristics and the withdrawal outcomes from the Cochrane Review. Please see the ‘acknowledgement’ section of this guideline.

The included studies differed in terms of the disease severity stated as an inclusion criterion as well as the treatment duration (see Table 70). The potential limitation of studies comparing interventions that act over different periods were noted(e.g., the faster acting clobetasol propionate and the slower acting calcipotriol), especially if the treatment duration chosen for the trial does not permit the maximum effect of the slower acting intervention to be observed.

Table 70. Disease severity inclusion criteria and treatment duration.

Table 70

Disease severity inclusion criteria and treatment duration.

8.2.2. Scalp psoriasis

8.2.2.1. Vitamin D or vitamin D analogue vs. placebo

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D or vitamin D analoguesPlaceboRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) - Calcipotriol OD (follow-up 4–8 weeks)
2
Jemec 2008
Green 1994
randomised trialsvery seriousaseriousbno serious indirectnessseriouscnone115/297 (38.7%)35/160 (21.9%)RR 2.12 (1.01 to 4.48)245 more per 1000 (from 2 more to 761 more)⊕○○○
VERY LOW
Patient’s assessment (clear/nearly clear) - Calcipotriol OD (follow-up 8 weeks)
1
Jemec 2008
randomised trialsseriouscno serious inconsistencyno serious indirectnessno serious imprecisionnone104/272 (38.2%)28/136 (20.6%)RR 1.86 (1.29 to 2.67)177 more per 1000 (from 60 more to 344 more)⊕⊕⊕○
MODERATE
Withdrawals due to adverse events - Calcipotriol OD (follow-up 4–8 weeks)
2
Jemec 2008
Green 1994
randomised trialsseriousdno serious inconsistencyno serious indirectnessvery seriousenone21/260 (8.1%)7/135 (5.2%)RR 1.44 (0.65 to 3.21)23 more per 1000 (from 18 fewer to 115 more)⊕○○○
VERY LOW
Withdrawals due to lack of efficacy - Calcipotriol OD (follow-up 4–8 weeks)
2
Jemec 2008
Green 1994
randomised trialsseriousfno serious inconsistencyno serious indirectnessseriousgnone19/258 (7.4%)18/146 (12.3%)RR 0.57 (0.31 to 1.06)53 fewer per 1000 (from 85 fewer to 7 more)⊕⊕○○
LOW
a

2/2 unclear allocation concealment; 1/2 high drop-out rate in both groups (21.0% of calcipotriol group and 22.1% of placebo); 1/2 unclear baseline comparability

b

Significant heterogeneity was present (I2 = 59%) that could not be explained in a clinically meaningful way by any of the pre-defined subgroups

c

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit to no clinically important benefit)

d

Unclear allocation concealment and high drop-out rate in both groups (21.0% of calcipotriol group and 22.1% of placebo)

e

Unclear allocation concealment and high drop-out rate in both groups (21.0% of calcipotriol group and 22.1% of placebo) in the trial weighted 94.8%

f

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

g

Unclear allocation concealment and high drop-out rate in both groups (21.0% of calcipotriol group and 22.1% of placebo) in the trial weighted 89.3%

h

Confidence interval ranges from clinically important effect to no effect

Evidence statements

In people with scalp psoriasis, topical calcipotriol once daily was statistically significantly better than placebo for:

  • Investigator’s assessment (clear/nearly clear) at 4–8 weeks [2 studies; 457 participants; very low quality evidence]128,168
  • Patient’s assessment (clear/nearly clear) at 8 weeks [1 study; 408 participants; moderate quality evidence]168

In people with scalp psoriasis, there was no statistically significant difference between topical calcipotriol once daily and placebo for:

  • Withdrawal due to adverse events at 4–8 weeks [2 studies; 395 participants; very low quality evidence]128,168
  • Withdrawal due to lack of efficacy at 4–8 weeks [2 studies; 404 participants; low quality evidence]128,168
Heterogeneity

For the outcome of investigators assessment of achieving clear/nearly clear status moderate heterogeneity was present between the results for the two studies128,168. This may have been partly a result of the small size of one of the studies128, but there were also other differences in the trials:

  • One study128 had a treatment duration of 4 weeks and used a calcipotriol solution, while the other168 had a treatment duration of 8 weeks and used the gel formulation. However, the results have not been separated as these differences were thought not to be a clinically feasible explanation for the inconsistency. The large effect estimate may have been caused by high risk of bias as this study had a small sample size and baseline demographics were not reported in this study. Nevertheless, both studies suggest that vitamin D or vitamin D analogues are clinically beneficial in terms of achieving clearance or near clearance compared with placebo treatment.

8.2.2.2. Potent corticosteroid vs. placebo

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCorticosteroid (potent)PlaceboRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) – betamethasone dipropionate OD or betamethasone valerate BD (follow-up 4–8 weeks)
2
Jemec 2008
Franz 1999
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone424/671 (63.2%)43/193 (22.3%)RR 2.81 (2.14 to 3.68)403 more per 1000 (from 254 more to 597 more)⊕⊕⊕○
MODERATE
Patient’s assessment (clear/nearly clear) – betamethasone dipropionate OD or betamethasone valerate BD (follow-up 4–8 weeks)
2
Jemec 2008
Franz 1999
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone419/671 (62.4%)38/193 (19.7%)RR 3.15 (2.35 to 4.21)423 more per 1000 (from 266 more to 632 more)⊕⊕⊕○
MODERATE
Withdrawals due to adverse events – betamethasone dipropionate OD or betamethasone valerate BD (follow-up 4–8 weeks)
2
Franz 1999
Jemec 2008
randomised trialsseriousbno serious inconsistencyno serious indirectnessno serious imprecisionnone6/630 (0.95%)7/170 (4.1%)RR 0.19 (0.06 to 0.55)33 fewer per 1000 (from 19 fewer to 39 fewer)⊕⊕⊕○
MODERATE
Withdrawals due to lack of efficacy - Betamethasone dipropionate OD (follow-up 8 weeks)
1
Jemec 2008
randomised trialsseriouscno serious inconsistencyno serious indirectnessno serious imprecisionnone9/518 (1.7%)16/122 (13.1%)RR 0.13 (0.06 to 0.29)114 fewer per 1000 (from 93 fewer to 123 fewer)⊕⊕⊕○
MODERATE
a

2/2 unclear allocation concealment; 1/2 (75.6% weighted) higher drop-out rate in placebo group (8.5% of active group and 22.1% of placebo); 1/2 (24.4% weighted) unclear blinding and dropout rates not given by group

b

2/2 unclear allocation concealment and 1/2 (100% weighted) higher drop-out rate in placebo group (8.5% of active group and 22.1% of placebo)

c

Unclear allocation concealment and higher drop-out rate in placebo group (8.5% of active group and 22.1% of placebo)

Evidence statements

In people with scalp psoriasis, topical potent corticosteroid treatment was statistically significantly better than placebo for:

  • Investigator’s assessment (clear/nearly clear) at 4–8 weeks for betamethasone dipropionate once daily or betamethasone valerate twice daily [2 studies; 864 participants; moderate quality evidence]108,168
  • Patient’s assessment at 4–8 weeks for betamethasone dipropionate once daily or betamethasone valerate twice daily (clear/nearly clear) [2 studies; 864 participants; moderate quality evidence] 108,168
  • Withdrawal due to adverse events at 4–8 weeks for betamethasone dipropionate once daily or betamethasone valerate twice daily [2 studies; 755 participants; moderate quality evidence]108,168
  • Withdrawal due to lack of efficacy at 8 weeks for betamethasone dipropionate once daily [1 study; 640 participants; moderate quality evidence] 168
Heterogeneity

No significant heterogeneity was detected between the studies despite differences in treatment duration (4108 vs 8168 weeks); intervention (betamethasone valerate108 vs dipropionate168); treatment frequency (once daily168 versus twice daily108) and treatment formulation (gel108 vs foam or lotion108).

One study108 found that foam was significantly more effective at achieving response (investigator’s assessment of clear/nearly clear) than lotion.

8.2.2.3. Very potent corticosteroid vs. placebo

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCorticosteroid (very potent)PlaceboRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) – clobetasol propionate OD/BD (follow-up 2–4 weeks)
4
Franz2000
Olsen 1991
Jarratt 2004
Sofen2011
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone290/449 (64.6%)27/339 (8%)RR 8.55 (5.88 to 12.43)601 more per 1000 (from 389 more to 910 more)⊕⊕⊕○
MODERATE
Patient’s assessment (clear/nearly clear) – clobetasol propionate BD (follow-up 2 weeks)
1
Franz2000
randomised trialsseriousbno serious inconsistencyno serious indirectnessno serious imprecisionnone77/125 (61.6%)4/63 (6.3%)RR 9.7 (3.72 to 25.3)552 more per 1000 (from 173 more to 1000 more)⊕⊕⊕○
MODERATE
Skin atrophy – clobetasol propionate OD/BD (follow-up 4 weeks)
2
Sofen2011 Jarratt 2004
randomised trialsseriouscno serious inconsistencyno serious indirectnessvery seriousdnone0/135 (0%)1/87 (1.1%)RR 0.33 (0.01 to 7.76)8 fewer per 1000 (from 11 fewer to 78 more)⊕○○○
VERY LOW
Withdrawals due to adverse events – clobetasol propionate OD/BD (follow-up 2–4 weeks)
4
Franz2000
Jarratt 2004
Sofen2011
Olsen 1991
randomised trialsseriouseno serious inconsistencyno serious indirectnessvery seriousdnone0/445 (0%)2/338 (0.59%)RR 0.34 (0.04 to 3.25)4 fewer per 1000 (from 6 fewer to 13 more)⊕○○○
VERY LOW
Withdrawals due to lack of efficacy – clobetasol propionate OD/BD (follow-up 2–4 weeks)
3
Olsen 1991
Franz2000
Jarratt 2004
randomised trialsseriousfno serious inconsistencyno serious indirectnessno serious imprecisionnone2/408 (0.49%)17/299 (5.7%)RR 0.12 (0.03 to 0.5)50 fewer per 1000 (from 28 fewer to 55 fewer)⊕⊕⊕○
MODERATE
a

4/4 unclear allocation concealment and 3/4 unclear blinding; 1/4 (22.9% weighted) unclear baseline comparability

b

Unclear allocation concealment, blinding and baseline comparability

c

2/2 unclear allocation concealment; 1/2 unclear blinding

d

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

e

4/4 unclear allocation concealment; 3/4 unclear blinding; 1/4 unclear baseline comparability

f

3/3 unclear allocation concealment and blinding; 1/3 unclear baseline comparability

Evidence statements

In people with scalp psoriasis, topical very potent corticosteroid treatment was statistically significantly better than placebo for:

  • Investigator’s assessment (clear/nearly clear) at 2–4 weeks for clobetasol propionate once or twice daily [4 studies; 788 participants; moderate quality evidence]109,166,292,377
  • Patient’s assessment (clear/nearly clear) at 2 weeks for clobetasol propionate twice daily [1 study; 188 participants; moderate quality evidence] 109
  • Withdrawal due to lack of efficacy at 2–4 weeks for clobetasol propionate once or twice daily [3 studies; 707 participants; moderate quality evidence]109,166,292

In people with scalp psoriasis, there was no statistically significant difference between topical very potent corticosteroid treatment and placebo for:

  • Skin atrophy at 4 weeks for clobetasol propionate once or twice daily [2 studies; 222 participants; very low quality evidence] 292,377
  • Withdrawal due to adverse events at 2–4 weeks for clobetasol propionate once or twice daily [4 studies; 783 participants; very low quality evidence] 292 109,166,377
Heterogeneity

No significant heterogeneity was detected between the studies despite differences in treatment duration (2109,292 vs 4166 weeks); treatment frequency (once daily166 versus twice daily109,292) and treatment formulation (solution292 vs shampoo166 vs foam or lotion109).

One study109 found that foam was more effective at achieving response (investigator’s assessment of clear/nearly clear) than solution (although no statistics were presented).

8.2.2.4. Combined product containing potent corticosteroid and vitamin D analogue (betamethasone dipropionate and calcipotriol) vs. placebo

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D and corticosteroid combinationPlaceboRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) (follow-up 8 weeks)
1
Tyring 2010
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone97/135 (80%)17/42 (50%)RR 1.77 (1.21 to 2.58)312 more per 1000 (from 85 more to 640 more)⊕⊕⊕○
MODERATE
Patient’s assessment (clear/nearly clear) (follow-up 8 weeks)
1
Tyring 2010
randomised trialsseriousano serious inconsistencyno serious indirectnessseriousbnone84/135 (62.2%)15/42 (35.7%)RR 1.74 (1.14 to 2.67)264 more per 1000 (from 50 more to 596 more)⊕⊕○○
LOW
Withdrawal due to adverse events (follow-up 8 weeks)
1
Tyring 2010
randomised trialsseriousano serious inconsistencyno serious indirectnessvery seriouscnone2/118 (1.7%)0/34 (0%)RR 1.47 (0.07 to 29.92)-⊕○○○
VERY LOW
a

Unclear allocation concealment

b

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit to no clinically important benefit)

c

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

Evidence statements

In people with scalp psoriasis, a combined product containing calcipotriol monohydrate and betamethasone dipropionate was statistically significantly better than placebo for:

  • Investigator’s assessment (clear/nearly clear) at 8 weeks [1 study; 177 participants; moderate quality evidence]405
  • Patient’s assessment (clear/nearly clear) at 8 weeks [1 study; 177 participants; low quality evidence]405

In people with scalp psoriasis, there was no statistically significant difference between a combined product containing calcipotriol monohydrate and betamethasone dipropionate and placebo for:

  • Withdrawal due to adverse events at 8 weeks [1 study; 152 participants; very low quality evidence]405
Subgroups and heterogeneity

One study405 performed a post-hoc subgroup analysis for the outcome of investigator’s assessment of clear/nearly clear to assess any difference between black/African-American and Hispanic/Latino subgroups of people with psoriasis. No significant difference was seen between the subgroups, although the results significantly favoured the combination over placebo in the Hispanic/Latino group (78 participants), but showed no significant difference in the Black/African-American group (99 participants).

8.2.2.5. Very potent corticosteroid vs. placebo for maintenance of remission

One study assessed the efficacy and safety of clobetasol propionate compared with placebo as a maintenance treatment for up to 6 months among those who had achieved clear, very mild or mild disease during a 4-week induction phase with once-daily clobetasol propionate. During the maintenance phase clobetasol propionate was used twice-weekly (3 days apart), but once daily dosing was permitted for up to 4 weeks if relapse occurred.

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsClobetasol propionatePlaceboRelative (95% CI)Absolute
Duration of remission (N still in remission) - 1 month (follow-up 1 month)
1
Poulin 2010
randomised trialsvery seriousano serious inconsistencyseriousbno serious imprecisionnone48/67 (71.6%)30/69 (43.5%)RR 1.65 (1.21 to 2.24)283 more per 1000 (from 91 more to 539 more)⊕○○○
VERY LOW
Duration of remission (N still in remission) - 2 months (follow-up 2 months)
1
Poulin 2010
randomised trialsvery seriousano serious inconsistencyseriousbno serious imprecisionnone41/67 (61.2%)20/69 (29%)RR 2.11 (1.39 to 3.2)322 more per 1000 (from 113 more to 638 more)⊕○○○
VERY LOW
Duration of remission (N still in remission) - 3 months (follow-up 3 months)
1
Poulin 2010
randomised trialsvery seriousano serious inconsistencyseriousbno serious imprecisionnone39/67 (58.2%)13/69 (18.8%)RR 3.09 (1.82 to 5.25)394 more per 1000 (from 154 more to 801 more)⊕○○○
VERY LOW
Duration of remission (N still in remission) - 4 months (follow-up 4 months)
1
Poulin 2010
randomised trialsvery seriousano serious inconsistencyseriousbno serious imprecisionnone34/67 (50.7%)11/69 (15.9%)RR 3.18 (1.76 to 5.75)348 more per 1000 (from 121 more to 757 more)⊕○○○
VERY LOW
Duration of remission (N still in remission) - 5 months (follow-up 5 months)
1
Poulin 2010
randomised trialsvery seriousano serious inconsistencyseriousbno serious imprecisionnone30/67 (44.8%)10/69 (14.5%)RR 3.09 (1.64 to 5.81)303 more per 1000 (from 93 more to 697 more)⊕○○○
VERY LOW
Duration of remission (N still in remission) - 6 months (follow-up 6 months)
1
Poulin 2010
randomised trialsvery seriousano serious inconsistencyseriousbno serious imprecisionnone27/67 (40.3%)8/69 (11.6%)RR 3.48 (1.7 to 7.1)288 more per 1000 (from 81 more to 707 more)⊕○○○
VERY LOW
Median time to relapse (follow-up 6 months)
1
Poulin 2010
randomised trialsvery seriousano serious inconsistencyseriousbseriouscnone6769-Placebo: 30.5 days

Clobetasol propionate: 141 days
⊕○○○
VERY LOW
Skin atrophy (follow-up 6 months)
1
Poulin 2010
randomised trialsvery seriousano serious inconsistencyno serious indirectnessvery seriousdnone1/67 (1.5%)0/69 (0%)RR 3.09 (0.13 to 74.5)-⊕○○○
VERY LOW
Withdrawals due to adverse events (follow-up 6 months)
1
Poulin 2010
randomised trialsvery seriousano serious inconsistencyno serious indirectnessvery seriousdnone2/60 (3.3%)0/52 (0%)RR 4.34 (0.21 to 88.48)-⊕○○○
VERY LOW
a

Unclear allocation concealment and blinding and higher drop-out rate in placebo group; patients in vehicle group received active treatment if relapse occurred during maintenance phase

b

Incorrect/less stringent definition of remission (at least mild on PGA)

c

No range given

d

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect Relapse data for this study is based on ITT analysis (worst case population; those who discontinued before relapse were considered as having relapse at the next visit)

Evidence statements

In people with scalp psoriasis, topical clobetasol propionate twice weekly maintenance treatment was statistically significantly better than placebo for:

  • Maintenance of remission at 1–6 months [1 study; 136 participants; very low quality evidence]315

In people with scalp psoriasis, there was no statistically significant difference between clobetasol propionate twice weekly maintenance treatment and placebo for:

  • Skin atrophy at 6 months [1 study; 136 participants; very low quality evidence]315
  • Withdrawal due to adverse events at 6 months [1 study; 112 participants; very low quality evidence]315

Evidence statement for individual study where no statistical analysis could be performed:

In people with psoriasis, clobetasol propionate twice weekly maintenance treatment was better than placebo for:

  • Median time-to-relapse among those who had achieved remission (maximum follow-up of 6 months) [1 study; 136 participants; very low quality evidence]315.

8.2.2.6. Vitamin D or vitamin D analogue vs. potent corticosteroid

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D or vitamin D analoguesCorticosteroid (potent)Relative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) – calcipotriol OD/BD vs betamethasone dipropionate OD or betamethasone valerate BD (follow-up 4–8 weeks)
3
Jemec 2008
Van de Kerkhof 2009
Klaber 1994
randomised trialsseriousavery seriousbno serious indirectnessno serious imprecisionnone362/794 (45.6%)874/1350 (64.7%)RR 0.69 (0.58 to 0.82)201 fewer per 1000 (from 117 fewer to 272 fewer)⊕○○○
VERY LOW
Patient’s assessment (clear/nearly clear) – calcipotriol OD/BD vs betamethasone dipropionate OD or betamethasone valerate BD (follow-up 4–8 weeks)
3
Jemec 2008
Van de Kerkhof 2009
Klaber 1994
randomised trialsseriousaseriouscno serious indirectnessno serious imprecisionnone368/794 (46.3%)856/1350 (63.4%)RR 0.71 (0.62 to 0.82)184 fewer per 1000 (from 114 fewer to 241 fewer)⊕⊕○○
LOW
Relapse rate - Calcipotriol BD vs betamethasone valerate BD (follow-up 4 weeks)
1
Klaber 1994
randomised trialsseriousdno serious inconsistencyseriouseseriousfnone75/99 (75.8%)102/129 (79.1%)RR 0.96 (0.83 to 1.1)32 fewer per 1000 (from 134 fewer to 79 more)⊕○○○
VERY LOW
Withdrawals due to adverse events – calcipotriol OD/BD vs betamethasone dipropionate OD or betamethasone valerate BD (follow-up 4–8 weeks)
3
Jemec 2008
Van de Kerkhof 2009
Klaber 1994
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone39/722 (5.4%)15/1246 (1.2%)RR 4.67 (2.57 to 8.48)44 more per 1000 (from 19 more to 90 more)⊕⊕⊕○
MODERATE
Withdrawals due to lack of efficacy – calcipotriol OD/BD vs betamethasone dipropionate OD or betamethasone valerate BD (follow-up 4–8 weeks)
3
Jemec 2008
Van de Kerkhof 2009
Klaber 1994
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone31/714 (4.3%)20/1251 (1.6%)RR 2.99 (1.73 to 5.19)32 more per 1000 (from 12 more to 67 more)⊕⊕⊕○
MODERATE
a

3/3 unclear allocation concealment; 2/3 unclear blinding; 1/3 higher dropout in vitamin D or vitamin D analogue group (21.0% in vitamin D or vitamin D analogue group and 8.5% in corticosteroid group)

b

Heterogeneity was present (I2 = 76%) that could not be explained by pre-defined subgroups (however, all studies showed the same direction of effect)

c

Heterogeneity was present (I2 = 65%) that could not be explained by pre-defined subgroups (however, all studies showed the same direction of effect)

d

Unclear allocation concealment and blinding

e

Surrogate outcome for duration of remission (defined as an increase in the total sign score to at least 50% of the score at the start of double-blind treatment)

f

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit to no clinically important benefit)

Evidence statements

In people with scalp psoriasis, topical potent corticosteroid treatment (betamethasone dipropionate once daily or betamethasone valerate twice daily) was statistically significantly better than topical vitamin D or vitamin D analogue (calcipotriol once or twice daily) for:

  • Investigator’s assessment (clear/nearly clear) at 4–8 weeks [3 studies; 2144 participants; very low quality evidence]168,189,408
  • Patient’s assessment (clear/nearly clear) at 4–8 weeks [3 studies; 2144 participants; low quality evidence]168,189,408
  • Withdrawals due to adverse events at 4–8 weeks [3 studies; 1968 participants; moderate quality evidence]168,189,408
  • Withdrawals due to lack of efficacy at 4–8 weeks [3 studies; 1965 participants; moderate quality evidence]168,189,408

In people with scalp psoriasis, there was no statistically significant difference between topical vitamin D analogue (calcipotriol twice daily) and potent corticosteroid (betamethasone valerate twice daily) for:

  • Relapse rate after a maximum follow-up of 4 weeks post-treatment [1 study; 228 participants; very low quality evidence]189
Heterogeneity

For the outcomes of investigator’s and patient’s assessment of achieving clear/nearly clear status high heterogeneity was present between the results for the three studies168,189,408. The heterogeneity was caused by the Jemec study in both cases, which gave a more favourable effect estimate for the potent corticosteroid. However, none of the pre-specified subgroups for investigation could explain this heterogeneity as there were no differences in study design or participant profile between the Jemec168 and van de Kerkhof408 studies. Although the Klaber study had a shorter treatment duration (4 vs 8 weeks), used twice rather than once daily dosing and betamethasone valerate solution rather than dipropionate gel, the result of this study was not the cause of the heterogeneity. However, the Jemec168 study did have a high drop-out in the calcipotriol arm, which may have biased the results. Nevertheless, both studies using betamethasone dipropionate suggest that there is precise evidence that potent corticosteroids are clinically beneficial in terms of achieving clearance or near clearance compared with vitamin D or vitamin D analogue treatment.

8.2.2.7. Vitamin D or vitamin D analogue vs. very potent corticosteroid

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D or vitamin D analoguesCorticosteroid (very potent)Relative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) - Calcipotriol (BD) vs clobetasol propionate (OD) (follow-up 4 weeks)
1
Reygagne 2005
randomised trialsseriousano serious inconsistencyseriousbseriouscnone21/75 (28%)38/76 (50%)RR 0.56 (0.37 to 0.86)220 fewer per 1000 (from 70 fewer to 315 fewer)⊕○○○
VERY LOW
Patient’s assessment (clear/nearly clear) - Calcipotriol (BD) vs clobetasol propionate (OD) (follow-up 4 weeks)
1
Reygagne 2005
randomised trialsseriousano serious inconsistencyseriousbseriouscnone23/75 (30.7%)36/76 (47.4%)RR 0.65 (0.43 to 0.98)166 fewer per 1000 (from 9 fewer to 270 fewer)⊕○○○
VERY LOW
Skin atrophy - Calcipotriol (BD) vs clobetasol propionate (OD) (follow-up 4 weeks)
1
Reygagne 2005
randomised trialsseriousdno serious inconsistencyseriousbvery seriouseNote that more cases of skin atrophy were present at baseline than week 4 and that in the clobetasol group it may only be 4 pts affected at different sites1/64 (1.6%)6/74 (8.1%)RR 0.19 (0.02 to 1.56)66 fewer per 1000 (from 79 fewer to 45 more)⊕○○○
VERY LOW
Withdrawals due to adverse events - Calcipotriol (BD) vs clobetasol propionate (OD) (follow-up 4 weeks)
1
Reygagne 2005
randomised trialsseriousano serious inconsistencyseriousbseriousfnone7/71 (9.9%)0/73 (0%)RR 15.42 (0.9 to 265)-⊕○○○
VERY LOW
a

Unclear allocation concealment; single blind (investigator); protocol violations included in ITT analysis; and relatively short duration of follow-up may produce an artificially high effect size in favour of the faster-acting clobetasol propionate

b

Different administration schedules for 2 groups: clobetasol once daily and washed out; calcipotriol twice daily and not washout out

c

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit/harm to no clinically important benefit/harm)

d

Unclear allocation concealment; single blind (investigator); protocol violations included in ITT analysis

e

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

f

Confidence interval ranges from clinically important effect to no effect

Evidence statements

In people with scalp psoriasis, topical very potent corticosteroid treatment (clobetasol propionate once daily) was statistically significantly better than topical vitamin D analogue (calcipotriol twice daily) for:

  • Investigator’s assessment (clear/nearly clear) at 4 weeks [1 study; 151 participants; very low quality evidence]330
  • Patient’s assessment (clear/nearly clear) at 4 weeks [1 study; 151 participants; very low quality evidence]330

In people with scalp psoriasis, there was no statistically significant difference between topical vitamin D analogue (calcipotriol twice daily) and very potent corticosteroid (clobetasol propionate once daily) for:

  • Skin atrophy at 4 weeks [1 study; 138 participants; very low quality evidence]330
  • Withdrawals due to adverse events at 4 weeks [1 study; 144 participants; very low quality evidence]330

8.2.2.8. Combined product containing vitamin D analogue and potent corticosteroid (betamethasone dipropionate and calcipotriol) vs. potent corticosteroid

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D and corticosteroid combinationPotent corticosteroidRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) - Combination OD vs. betamethasone dipropionate OD (follow-up 8 weeks)
2
Jemec 2008
van de Kerkhof 2009
randomised trialsseriousano serious inconsistencyno serious indirectnessseriousbnone773/1180 (65.5%)699/1118 (62.5%)RR 1.12 (1.05 to 1.18)75 more per 1000 (from 31 more to 113 more)⊕⊕○○
LOW
Patient’s assessment (clear/nearly clear) - Combination OD vs. betamethasone dipropionate OD (follow-up 8 weeks)
3
Buckley 2008
Jemec 2008
van de Kerkhof 2009
randomised trialsseriouscno serious inconsistencyno serious indirectnessseriousbnone866/1216 (71.2%)776/1228 (63.2%)RR 1.13 (1.07 to 1.19)82 more per 1000 (from 44 more to 120 more)⊕⊕○○
LOW
Withdrawals due to adverse events - Combination OD vs. betamethasone dipropionate OD (follow-up 8 weeks)
3
Buckley 2008
Jemec 2008
van de Kerkhof 2009
randomised trialsseriouscseriousdno serious indirectnessvery seriousenone13/1107 (1.2%)15/1122 (1.3%)RR 0.88 (0.42 to 1.85)2 fewer per 1000 (from 8 fewer to 11 more)⊕○○○
VERY LOW
Withdrawals due to lack of efficacy - Combination OD vs. betamethasone dipropionate OD (follow-up 8 weeks)
3
Buckley 2008
Jemec 2008
van de Kerkhof 2009
randomised trialsseriouscno serious inconsistencyno serious indirectnessseriousfnone9/1103 (0.82%)20/1127 (1.8%)RR 0.47 (0.22 to 1.01)9 fewer per 1000 (from 14 fewer to 0 more)⊕⊕○○
LOW
a

2/2 unclear allocation concealment; 1/2 unclear blinding

b

Serious imprecision according to GDG discussion (confidence interval ranges from clinically important benefit to no clinically important benefit)

c

3/3 unclear allocation concealment; 2/3 unclear blinding

d

No heterogeneity detected statistically due to very wide confidence intervals but studies show different directions of effect

e

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

f

Confidence interval ranges from clinically important effect to no effect

Evidence statements

In people with scalp psoriasis, a combined product containing calcipotriol monohydrate and betamethasone dipropionate was statistically significantly better than potent corticosteroid alone (betamethasone dipropionate once daily) for:

  • Investigator’s assessment (clear/nearly clear) at 8 weeks [2 studies; 1472 participants; low quality evidence]48,168,408
  • Patient’s assessment (clear/nearly clear) [3 studies; 2226 participants; low quality evidence] 48,168,408

In people with scalp psoriasis, there was no statistically significant difference between a combined product containing calcipotriol monohydrate and betamethasone dipropionate and potent corticosteroid alone (betamethasone dipropionate once daily) for:

  • Withdrawal due to adverse events at 8 weeks [3 studies; 2229 participants; very low quality evidence]48,168,408
  • Withdrawal due to lack of efficacy at 8 weeks [3 studies; 2230 participants; low quality evidence]48,168,408
Heterogeneity

No significant heterogeneity was detected between the studies and all had the same treatment duration, formulation and frequency as well as the same inclusion criteria in terms of disease severity.

8.2.2.9. Combined product containing vitamin D analogue and potent corticosteroid (betamethasone dipropionate and calcipotriol) vs. vitamin D or vitamin D analogue

One study228 assessed long-term (52 weeks) treatment for this comparison. This study used a once daily administration schedule as required by the participants and the mean treatment duration was 44 weeks and 37 weeks for the combination and vitamin D or vitamin D groups, respectively (mean weekly weight used: 10.6g in two compound group and 12.8g in calcipotriol group; mean weight used over whole study period 470.8g and 440.0g, respectively).

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVitamin D and corticosteroid combinationVitamin D or vitamin D analogueRelative (95% CI)Absolute
Investigator’s assessment (clear/nearly clear) – combination OD vs calcipotriol OD/BD (follow-up 8 weeks)
3
Kragballe2009
Jemec 2008
van de Kerkhof 2009
randomised trialsvery seriousaseriousbno serious indirectnessno serious imprecisionnone915/1315 (69.6%)257/663 (38.8%)RR 1.83 (1.52 to 2.20)322 more per 1000 (from 202 more to 465 more)⊕○○○
VERY LOW
Patient’s assessment (clear/nearly clear) - combination OD gel vs calcipotriol OD gel (follow-up 8 weeks)
2
Jemec 2008
van de Kerkhof 2009
randomised trialsvery seriouscno serious inconsistencyno serious indirectnessno serious imprecisionnone766/1108 (69.1%)232/558 (41.6%)RR 1.66 (1.5 to 1.85)274 more per 1000 (from 208 more to 353 more)⊕⊕○○
LOW
Patient’s assessment (clear/nearly clear) - combination OD gel vs calcipotriol BD solution (follow-up 8 weeks)
1
Kragballe2009
randomised trialsvery seriousdno serious inconsistencyno serious indirectnessno serious imprecisionnone170/207 (82.1%)36/105 (34.3%)RR 2.4 (1.82 to 3.15)480 more per 1000 (from 281 more to 737 more)⊕⊕○○
LOW
Skin atrophy - combination OD vs calcipotriol BD (follow-up 8 weeks)
1
Kragballe2009
randomised trialsvery seriousdno serious inconsistencyno serious indirectnessno serious imprecisionnone0/207 (0%)0/105 (0%)not poolednot pooled⊕⊕○○
LOW
Skin atrophy - combination OD vs calcipotriol OD (follow-up 52 weeks)
1
Luger 2008
randomised trialsseriouseno serious inconsistencyno serious indirectnessno serious imprecisionnone0/429 (0%)0/440 (0%)not poolednot pooled⊕⊕⊕○
MODERATE
Relapse rate - combination OD vs calcipotriol BD (follow-up 8 weeks)
1
Kragballe2009
randomised trialsvery seriousfno serious inconsistencyseriousgserioushnone73/135 (54.1%)10/29 (34.5%)RR 1.57 (0.93 to 2.65)197 more per 1000 (from 24 fewer to 569 more)⊕○○○
VERY LOW
Median time to relapse - combination OD vs calcipotriol BD
1
Kragballe2009
randomised trialsvery seriousfno serious inconsistencyno serious indirectnessseriousinone13529Combination: 35 days

Vitamin D analogue: 58 days
⊕○○○
VERY LOW
Withdrawals due to adverse events - combination OD vs calcipotriol OD/BD (follow-up 8 weeks)
3
Kragballe2009
Jemec 2008
van de Kerkhof 2009
randomised trialsvery seriousano serious inconsistencyno serious indirectnessno serious imprecisionnone14/1204 (1.2%)37/582 (6.4%)RR 0.18 (0.1 to 0.33)52 fewer per 1000 (from 43 fewer to 57 fewer)⊕⊕○○
LOW
Withdrawals due to lack of efficacy - combination OD vs calcipotriol OD (follow-up 8 weeks)
2
Jemec 2008
van de Kerkhof 2009
randomised trialsvery seriousjvery seriouskno serious indirectnessvery seriousjnone9/1009 (0.89%)27/490 (5.5%)RR 0.16 (0.02 to 1.35)46 fewer per 1000 (from 54 fewer to 19 more)⊕○○○
VERY LOW
Withdrawals due to adverse events - combination OD vs calcipotriol OD (follow-up 52 weeks)
1
Luger 2008
randomised trialsseriouseno serious inconsistencyno serious indirectnessno serious imprecisionnone9/346 (2.6%)44/309 (14.2%)RR 0.18 (0.09 to 0.37)117 fewer per 1000 (from 90 fewer to 130 fewer)⊕⊕⊕○
MODERATE
Withdrawals due to lack of efficacy - combination OD vs calcipotriol OD (follow-up 52 weeks)
1
Luger 2008
randomised trialsseriouseno serious inconsistencyno serious indirectnessno serious imprecisionnone14/351 (4%)51/316 (16.1%)RR 0.25 (0.14 to 0.44)121 fewer per 1000 (from 90 fewer to 139 fewer)⊕⊕⊕○
MODERATE
a

3/3 unclear allocation concealment; 1/3 (48.2% weighted) unclear blinding; 1/3 single blind (investigator); 2/3 higher dropout with vitamin D or vitamin D analogue

b

Heterogeneity was present (I2 = 64%) that could not be explained by pre-defined subgroups (however, all studies showed the same direction of effect and the p-value for chi squared was >0.05)

c

2/2 unclear allocation concealment; 1/2 single blind (investigator); 1/2 higher dropout rate in vitamin D or vitamin D analogue group (22.1% vs 11.3% in combination group)

d

Unclear allocation concealment; single blind (investigator); higher dropout in vitamin D or vitamin D analogue group (21.9% vs 8.2% in combination group)

e

Unclear allocation concealment

f

Unclear allocation concealment; single blind (investigator); higher dropout in vitamin D or vitamin D analogue group (21.9% vs 8.2% in combination group); also, unclear baseline comparability as only includes those in each group who achieved remission; therefore, there are also fewer participants in the vitamin D or vitamin D analogue group

g

Surrogate outcome for duration of remission

h

Confidence interval ranges from clinically important effect to no effect

i

No range given

j

2/2 unclear allocation concealment; 1/2 unclear blinding; 1/2 higher dropout rate in vitamin D or vitamin D analogue group (22.1% vs 11.3% in combination group)

k

Heterogeneity was present (I2 = 80%) that could not be explained by pre-defined subgroups (however, all studies showed the same direction of effect)

l

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

Evidence statements

In people with scalp psoriasis, a combined product containing calcipotriol monohydrate and betamethasone dipropionate was statistically significantly better than vitamin D analogue alone (calcipotriol once or twice daily) for:

  • Investigator’s assessment (clear/nearly clear) at 8 weeks [3 studies; 1978 participants; very low quality evidence]168,199,408
  • Patient’s assessment (clear/nearly clear) at 8 weeks [3 studies; 1978 participants; low quality evidence]168,199,408
  • Withdrawals due to adverse events at 8 weeks [3 studies; 1786 participants; low quality evidence]168,199,408
  • Withdrawals due to adverse events at 52 weeks [1 study; 655 participants; moderate quality evidence]228
  • Withdrawals due to lack of efficacy at 52 weeks [1 study; 667 participants; moderate quality evidence]228

In people with scalp psoriasis, there were no events with either a combined product containing calcipotriol monohydrate and betamethasone dipropionate or vitamin D analogue alone (calcipotriol once or twice daily) for:

  • Skin atrophy at 8 or 52 weeks [2 studies; 312 and 869 participants; low to moderate quality evidence]199,228

In people with scalp psoriasis, there was no statistically significant difference between a combined product containing calcipotriol monohydrate and betamethasone dipropionate and topical vitamin D analogue alone for:

  • Relapse rate at 8 weeks post-treatment for the combined product compared with calcipotriol twice daily [1 study; 164 participants; very low quality evidence]199
  • Withdrawals due to lack of efficacy at 8 weeks for the combined product compared with calcipotriol once daily [2 studies; 1499 participants; very low quality evidence]168,408

Evidence statement for an individual study where no statistical analysis could be performed comparing a combined product containing calcipotriol monohydrate and betamethasone dipropionate and vitamin D analogue alone for scalp psoriasis:

  • The median time to relapse was longer with calcipotriol twice daily than with the combination treatment after a maximum follow-up of 8 weeks post-treatment [1 study; 164 participants; very low quality evidence]199
Heterogeneity

For the outcome of investigator’s assessment of achieving clear/nearly clear status high heterogeneity was present between the results for the three studies168,199,408. The heterogeneity was caused by the van de Kerkhof study, which gave an effect estimate that was slightly less favourable for the combination. However, none of the pre-specified subgroups for investigation could explain this heterogeneity as there were no differences in study design or participant profile between the Jemec168 and van de Kerkhof408 studies. Although the Kragballe study199 used twice rather than once daily dosing of calcipotriol, the result of this study was not the cause of the heterogeneity. Differences in risk of bias did not explain the inconsistency either. Nevertheless, all three studies demonstrate that there is precise evidence that the combination is clinically beneficial in terms of achieving clearance or near clearance compared with vitamin D or vitamin D analogue treatment alone.

For the patient’s assessment of achieving clear/nearly clear status high heterogeneity was present between the results for the three studies168,199,408. This was explained by creating subgroups based on the treatment formulation, as the Kragballe 2009199 study used a gel for the combination arm and a solution for the calcipotriol arm, which resulted in a greater effect estimate in favour of the combination treatment. Note that although the treatment frequency was also different in the Kragballe 2009199 study (twice daily calcipotriol compared with once daily in the other two studies168,408) this is not a clinically relevant explanation for the heterogeneity as the study with twice daily calcipotriol199 favours the combination more highly.

8.2.2.10. Very potent corticosteroid vs. coal tar polytherapy

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsVery potent corticosteroidCoal tar polytherapyRelative (95% CI)Absolute
Skin atrophy - Clobetasol propionate OD vs polytar twice weekly (follow-up 4 weeks)
1
Griffiths2006A
randomised trialsvery seriousano serious inconsistencyno serious indirectnessno serious imprecisionnone0/121 (0%)0/41 (0%)not poolednot pooled⊕⊕○○
LOW
Withdrawal due to adverse events - Clobetasol propionate OD vs polytar twice weekly (follow-up 4 weeks)
1
Griffiths2006A
randomised trialsvery seriousano serious inconsistencyno serious indirectnessvery seriousbnone1/121 (0.83%)0/41 (0%)RR 1.03 (0.04 to 24.87)-⊕○○○
VERY LOW
a

Unclear allocation concealment and blinding; unclear dropout rates; higher proportion of males in the tar group (65.9% vs 48.8%)

b

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

Evidence statements

In people with scalp psoriasis, there were no events with either very potent corticosteroid (clobetasol propionate once daily) or coal tar polytherapy twice weekly for:

  • Skin atrophy at 4 weeks [1 study; 162 participants; low quality evidence]130

In people with scalp psoriasis, there was no statistically significant difference between very potent corticosteroid (clobetasol propionate once daily) and coal tar polytherapy twice weekly for:

  • Withdrawal due to adverse events at 4 weeks [1 study; 162 participants; very low quality evidence]130

8.2.2.11. Vitamin D analogue vs. coal tar polytherapy

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCalcipotriolCoal tar polytherapyRelative (95% CI)Absolute
Investigators assessment (at least moderate improvement) - Calcipotriol BD vs. coal tar polytherapy OD (follow-up 8 weeks)
1
McKinnon2000
randomised trialsvery seriousano serious inconsistencyseriousbno serious imprecisionnone120/210 (57.1%)79/213 (37.1%)RR 1.54 (1.25 to 1.9)200 more per 1000 (from 93 more to 334 more)⊕○○○
VERY LOW
Withdrawals due to adverse events - Calcipotriol BD vs. coal tar polytherapy OD (follow-up 8 weeks)
1
McKinnon2000
randomised trialsvery seriousano serious inconsistencyno serious indirectnessno serious imprecisionnone35/230 (15.2%)16/215 (7.4%)RR 2.04 (1.17 to 3.59)77 more per 1000 (from 13 more to 193 more)⊕⊕○○
LOW
a

Unclear allocation concealment; unblinded; high dropout rate (30.3% in vitamin D analogue and 29.1% in tar group)

b

Incorrect definition of response (at least moderate improvement)

Evidence statements

In people with scalp psoriasis, vitamin D analogue (calcipotriol twice daily) was statistically significantly better than coal tar polytherapy (once daily) for:

  • Investigator’s assessment (at least moderate improvement) at 8 weeks [1 study; 423 participants; very low quality evidence]245

In people with scalp psoriasis, coal tar polytherapy (once daily) was statistically significantly better than vitamin D analogue (calcipotriol twice daily) for:

  • Withdrawal due to adverse events at 8 weeks [1 study; 445 participants; low quality evidence]245

8.2.3. Time to remission or maximum effect for scalp psoriasis

8.2.3.1. Vitamin D or vitamin D analogues

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCalcipotriol
Time-to-absent/very mild disease (follow-up 1 week)
1
Jemec2011 (pooled data from Jemec2008 & van de Kerkhof 2009)
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone558Patients achieving absent or very mild disease

Week 1: 54/545 (10.0%)
⊕⊕○○
LOW
Time-to-absent/very mild disease (follow-up 2–8 weeks)
1
Jemec 2008
observational studiesano serious risk of biascno serious inconsistencyno serious indirectnessno serious imprecisionnone272Patients achieving absent or very mild disease

Week 2: 51 (18.8%)

Week 4: 64 (23.5%)

Week 8: 100 (36.8%)
⊕⊕○○
LOW
Time-to-absent/very mild disease (follow-up 2–8 weeks)
1
van de Kerhof2009
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone286Patients achieving absent or very mild disease

Week 2: 45 (15.7%)

Week 4: 74 (25.9%)

Week 8: 124 (43.4%)
⊕⊕○○
LOW
Time-to-absent/very mild disease (follow-up 2–8 weeks)
1
Kragballe 2009
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone105Patients achieving absent or very mild disease

Week 2: 11 (10.5%)

Week 4: 19 (18.1%)

Week 8: 33 (31.4%)
⊕⊕○○
LOW
Mean time to maximum response (change in TSS) (follow-up 24 weeks)
1
McKinnon 2000
observational studiesano serious risk of biasbno serious inconsistencyseriousdvery seriousenone238Based on change in TSS maximum effect was not reached by the end of 8 weeks comparative phase

Over the long-term treatment phase based on graphical representation of change in TSS most of the improvement is achieved by 12 weeks, with only slight further improvement up to 24 weeks (approximately 1 point reduction on TSS over 12 weeks)
⊕○○○
VERY LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Unclear allocation concealment may have biased patient selection for this intervention and there was a high rate of dropout (21.0%)

d

Incorrect outcome measure

e

Interpreted from graphical representation

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for topical vitamin D or vitamin D analogues (no statistical analysis could be performed).

In people with scalp psoriasis, the time to remission when using calcipotriol varied between studies:

  • Proportion achieving remission by 8 weeks ranged from 31.4 to 43.4% [3 studies; 663 participants; low quality evidence]168,199,408
  • The continued increase in responders between 4 and 8 weeks ranged from 13.3–17.5% [3 studies; 663 participants; low quality evidence]168,199,408
  • Some people (10%) achieved remission by 1 week [1 study; 558 participants; low quality evidence]169
  • Of those who achieved remission by the end of the trial (8 weeks), 57.6–64.0% had responded by week 4 based on investigators assessment [3 studies; 663 participants; low quality evidence]168,199,408
  • Graphical representation of longer-term data demonstrated that the majority of the improvement in TSS score is achieved by 12 weeks, with only slight further improvement up to 24 weeks (approximately 1 point reduction on TSS over the second 12 weeks) [1 study; 238 participants; very low quality evidence]245
Summary

The evidence suggests that maximum response is not achieved in all patients by 8 weeks, with the response rate still increasing at this time point 168,199,408, and one study245 suggests that 12 weeks may represent the time at which maximum response is achieved.

8.2.3.2. Potent corticosteroids

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsBetamethasone dipropionate
Time-to-absent/very mild disease (follow-up 1 week)
1
Jemec2011 (pooled data from Jemec2008 & van de Kerkhof 2009)
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone1118Patients achieving absent or very mild disease

Week 1: 262 (24.1%)
⊕⊕○○
LOW
Time-to-absent/very mild disease (follow-up 2–8 weeks)
1
Jemec 2008
observational studiesano serious risk of biascno serious inconsistencyno serious indirectnessno serious imprecisionnone562Patients achieving absent or very mild disease

Week 2: 262 (47.1%)

Week 4: 304 (54.7%)

Week 8: 356 (64.0%)
⊕⊕○○
LOW
Time-to-absent/very mild disease (follow-up 2–8 weeks)
1
van de Kerhof2009
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone556Patients achieving absent or very mild disease

Week 2: 216 (38.4%)

Week 4: 287 (51.1%)

Week 8: 343 (61.0%
⊕⊕○○
LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Unclear allocation concealment may have biased patient selection for this intervention and there was a high rate of dropout (21.0%)

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for topical potent corticosteroids (no statistical analysis could be performed).

In people with scalp psoriasis, the time to remission when using betamethasone dipropionate varied between studies:

  • Proportion achieving remission by 8 weeks ranged from 61.0 to 64.0% [2 studies; 1118 participants; low quality evidence]168,408
  • The continued increase in responders between 4 and 8 weeks ranged from 9.3–9.9% [2 studies; 1118 participants; low quality evidence]168,408
  • Some people (24.1%) achieved remission by 1 week [1 study; 262 participants; low quality evidence]169
  • Of those who achieved remission by the end of the trial (8 weeks), 63.0–73.6% had responded by week 2 and 83.7–85.4% by week 4 based on investigators assessment [2 studies; 1118 participants; low quality evidence]168,408
Summary

The evidence suggests that maximum response is not achieved in all patients by 8 weeks, with the response rate still increasing at this time point 168,408. However, the majority of those who will respond within 8 weeks had done so by week 4.

8.2.3.3. Very potent corticosteroids

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsClobetasol propionate
Time-to-clear/nearly clear disease (follow-up 4 weeks)
1
Sofen 2011
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone81Patients achieving clear/nearly clear disease

Week 2: 33/41 (80.5%)

Week 4: 35/41 (85.4%)
⊕⊕○○
LOW
Mean time to maximum response (TSS) (follow-up 4 weeks)
1
Reygagne 2005
observational studiesano serious risk of biasbno serious inconsistencyseriouscvery seriousdnone232Graphical representation of mean TSS over time shows a large effect by week 2 which begins to slow between weeks 2–4, with continued gradual reduction in mean TSS)⊕○○○
VERY LOW
Mean time to maximum response (TSS) (follow-up 4 weeks)
1
Jarratt 2004
observational studiesano serious risk of biasbno serious inconsistencyseriouscvery seriousdnone95Score for TSS decreased rapidly from baseline to week four, but did not reach maximum effect (2-wk post-treatment follow-up showed a slight increase in TSS)⊕○○○
VERY LOW
Mean time to maximum response (TSS) (follow-up 2 weeks)
1
Franz 2000
observational studiesano serious risk of biasbno serious inconsistencyseriouscvery seriousdnone125Maximum effect was not reached for scaling, plaque thickness, pruritus and erythema scores by 14 days; the mean severity score increased during the 14 days following removal of treatment⊕○○○
VERY LOW
Mean time to maximum response (PAGI) (follow-up 4 weeks)
1
Griffiths 2006A
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessvery seriousdnone121Continued improvement was seen between weeks 2 and 4 based on improvement in participants’ global assessment of improvement from baseline⊕○○○
VERY LOW
Mean time to remission (PGA) (follow-up 4 weeks)
1
Poulin 2010
observational studiesano serious risk of biasbno serious inconsistencyseriouseno serious imprecisionnone6789% (141/168) of those entered into the induction phase achieved clear, mild or very⊕○○○
VERY LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Incorrect outcome measure

d

Interpreted from graphical representation

e

Incorrect definition of response (at least mild on PGA)

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for topical very potent corticosteroids (no statistical analysis could be performed).

  • In people with scalp psoriasis, the time to remission when using clobetasol propionate varied between studies:
  • Proportion achieving remission by 4 weeks was 85.4% [1 study; 81 participants; low quality evidence]377
  • The continued increase in responders between 2 and 4 weeks was 4.9% [1 study; 81 participants; low quality evidence]377
  • Of those who achieved remission by the end of the trial (4 weeks), 94.3% had responded by week 2 [1 study; 81 participants; low quality evidence]377
  • Mean TSS shows a rapid effect over the first 2 weeks of treatment, but has not reached a maximum effect by week 2 or 4 [3 studies; 452 participants; very low quality evidence]109,166,330
  • Patient’s global improvement scores show that continued improvement was seen between weeks 2 and 4 [1 study; 121 participants; very low quality evidence]130
  • Investigator’s global assessment of response (clear, mild or very mild disease) showed that 89% achieved remission by week 4 [1 study; 67 participants; very low quality evidence]315.
Summary

The evidence suggests that maximum response is not achieved in all patients by 2 or 4 weeks, with the response rate still increasing at this time point109,130,166,315,330.

8.2.3.4. Combined product containing potent corticosteroid and vitamin D analogue (betamethasone dipropionate and calcipotriol monohydrate)

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCombined betamethasone dipropionate and calcipotriol
Time-to-absent/very mild disease (follow-up 1 week)
1
Jemec2011 (pooled data from Jemec2008 & van de Kerkhof 2009)
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone1108Patients achieving absent or very mild disease

Week 1: 331 (30.6%)
⊕⊕○○
LOW
Time-to-absent/very mild disease (follow-up 2–8 weeks)
1
Jemec 2008
observational studiesano serious risk of biascno serious inconsistencyno serious indirectnessno serious imprecisionnone541Patients achieving absent or very mild disease

Week 2: 311 (57.5%)

Week 4: 362 (66.9%)

Week 8: 385 (71.2%)
⊕⊕○○
LOW
Time-to-absent/very mild disease (follow-up 2–8 weeks)
1
van de Kerhof2009
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone567Patients achieving absent or very mild disease

Week 2: 278 (49.0%)

Week 4: 311 (54.9%)

Week 8: 388 (68.4%)
⊕⊕○○
LOW
Time-to-absent/very mild disease (follow-up 2–8 weeks)
1
Kragballe 2009
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnone207Patients achieving absent or very mild disease

Week 2: 125 (60.4%)

Week 4: 114 (55.1%)

Week 8: 142 (68.6%)
⊕⊕○○
LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Unclear allocation concealment may have biased patient selection for this intervention and there was a high rate of dropout (21.0%)

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for topical combination therapies (no statistical analysis could be performed).

In people with scalp psoriasis, the time to remission when using a combined product containing betamethasone dipropionate and calcipotriol varied between studies:

  • Proportion achieving remission by 8 weeks ranged from 68.4 to 71.2% [3 studies; 1315 participants; low quality evidence]168,199,408
  • The continued increase in responders between 4 and 8 weeks ranged from 4.3–13.5% [3 studies; 1315 participants; low quality evidence]168,199,408
  • Some people (30.6%) achieved remission by 1 week [1 study; 1108 participants; low quality evidence]169,170
  • Of those who achieved remission by the end of the trial (8 weeks), 71.6–88.0% had responded by week 2 and 80.2–94.0% by week 4 based on investigators assessment [3 studies; 1315 participants; low quality evidence]168,199,408
Summary
  • The evidence suggests that maximum response is not achieved in all patients by 8 weeks, with the response rate still increasing at this time point168,199,408. However, the majority of those who will respond within 8 weeks had done so by weeks 2–4.

8.2.3.5. Coal tar

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsCoal tar
Mean time to maximum response (change in TSS) (follow-up 8 weeks)
1
McKinnon 2000
observational studiesano serious risk of biasbno serious inconsistencyseriouscvery seriousdnone237Based on change in TSS maximum effect was not reached by the end of the study period (8 weeks)⊕○○○
VERY LOW
Mean time to maximum response (patients’ assessment) (follow-up 4 weeks)
1
Griffiths 2006A
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessvery seriousdnone41A very small amount of continued improvement was seen between weeks 2 and 4 based on change in participants’ global assessment of improvement from baseline⊕○○○
VERY LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Incorrect outcome measure

d

Interpreted from graphical representation

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for topical coal tar therapies (no statistical analysis could be performed).

In people with scalp psoriasis, the time to remission when using coal tar varied between studies:

  • Mean change in TSS showed that a maximum effect was not reached by week 8 [1 study; 237 participants; very low quality evidence]245
  • Patient’s assessment of global improvement showed that very slight continued improvement was seen between weeks 2 and 4 [1 study; 41 participants; very low quality evidence]130
Summary

The evidence suggests that maximum response based on TSS is not achieved in all patients by 8 weeks, with the response rate still increasing at this time point245, although the results at 4 weeks suggest that response based on patient’s global assessment may begin to plateau between 2 and 4 weeks130.

8.2.4. Network meta-analysis – scalp psoriasis

Based on the results of conventional meta-analyses of direct evidence alone, it can be difficult to determine which intervention is most effective in the treatment of chronic plaque psoriasis. The challenge of interpretation arises for two reasons:

  • Some pairs of alternative strategies have not been directly compared in a randomised controlled trial (for example, very potent corticosteroid vs a combined product containing vitamin D analogue and potent corticosteroid)
  • There are frequently multiple overlapping comparisons (for example vitamin D or vitamin D analogue vs potent corticosteroid, vitamin D or vitamin D analogue vs a combined product containing vitamin D analogue and potent corticosteroid and potent corticosteroid vs a combined product containing vitamin D analogue and potent corticosteroid) that could potentially give inconsistent estimates of effect.

To overcome these problems, a hierarchical Bayesian network meta-analysis (NMA) was performed. This type of analysis allows for the synthesis of data from direct and indirect comparisons and allows for the ranking of different interventions in order of efficacy, defined as the achievement of clearance or near clearance. A network meta-analysis also provides estimates of effect (with 95% credible interval) for each intervention compared to one another and compared to a single baseline risk. These estimates provide a useful and coherent clinical summary of the results and facilitate the formation of recommendations based on the best available evidence. Furthermore, these estimates were used to parameterise treatment effectiveness of the topical therapies in the original cost-effectiveness modelling outlined in section 8.2.5. For details on the methods, results and interpretation of the network meta-analyses, see Appendix L.

The inclusion criteria for and intervention compared in the NMA were the same as in the review of direct evidence (Section 8.2.1). A class effect was still assumed, but in order to reduce heterogeneity in the network of evidence, interventions were broken down by treatment frequency from the outset. In other words, once daily vitamin D or vitamin D analogue and twice daily vitamin D or vitamin D analogue were considered separate comparators in the NMA. Placebo/vehicle delivered once daily was also considered separately from twice daily placebo/vehicle.

The outcomes considered as part of the NMA were restricted to those measuring response:

  • Clear/nearly clear or marked improvement (at least 75% improvement) on Investigator’s assessment of overall global improvement (IAGI) or clear/nearly clear/minimal (not mild) on Physician’s Global Assessment (PGA)

Unfortunately, the network of evidence for the outcome of clear/nearly clear or marked improvement (at least 75% improvement) on the Patient’s assessment of overall global improvement (PAGI) or clear/nearly clear/minimal (not mild) on Patient’s Global Assessment was not connected such that an analysis could be performed.

8.2.4.1. Results of NMA for investigator assessed outcome: clear/nearly clear (IAGI/PGA)

A total of 13 studies108,109,128,166,168,189,199,245,292,330,377,405,408 from the original evidence review met the inclusion criteria for the network. Based on the GRADE quality ratings from the review of direct comparisons (section 8.2.2), the evidence included in the network meta-analysis ranges in quality from very low to moderate.

Figure 6 presents all the interventions included in the NMA as well as shows where there is direct evidence for a particular comparison and the number of studies that have included that comparison. For example, there are 3 studies reporting the outcome ‘clear’ or ‘nearly clear’ as measured by IAGI or PGA for the comparison of twice daily vehicle/placebo and twice daily very potent corticosteroid. The diagram also highlights where there are gaps in the direct evidence. For example, there are no studies comparing a combined product containing vitamin D or vitamin D analogue and potent corticosteroid to very potent corticosteroid.

Figure 6. Clear or nearly clear – IAGI and PGA.

Figure 6

Clear or nearly clear – IAGI and PGA. Note: Solid lines indicate direct head-to-head comparisons and the colour indicates the number of trials per comparison included in the analysis.

The results of the network meta-analysis in terms of the relative risk of each intervention compared to twice daily vehicle/placebo are presented in Table 71. It also gives a probability that the intervention is the most effective overall.

Table 71. Relative risks of clear/nearly clear on IAGI/PGA for all interventions compared to twice daily vehicle/placebo.

Table 71

Relative risks of clear/nearly clear on IAGI/PGA for all interventions compared to twice daily vehicle/placebo.

Evidence statements

Results of the network meta-analysis of randomised controlled trials indicate that in the treatment of patients with scalp psoriasis the following interventions are statistically significantly more effective than twice daily vehicle/placebo at inducing clearance/near clearance as measured by the investigator or physician (IAGI/PGA):

  • Once and twice daily very potent corticosteroid
  • Once and twice daily potent corticosteroid
  • Once and twice daily vitamin D or vitamin D analogue
  • Once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate

Results of the network meta-analysis of randomised controlled trials indicate that in the treatment of patients with scalp psoriasis there is no statistically significant difference between once daily coal tar polytherapy and twice daily placebo in terms of achieving clearance/near clearance as measured by the investigator or physician (IAGI/PGA).

Results of the network meta-analysis of scalp psoriasis treatments indicate that there are very few comparisons between active treatments for which the treatment effect reaches statistical significance. A few exceptions include:

  • Once daily potent corticosteroid is more effective than once daily vitamin D or vitamin D analogue
  • Once and twice daily very potent corticosteroids are more effective than once and twice daily vitamin D or vitamin D analogue and once daily coal tar polytherapy
  • Once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate is more effective than once vitamin D or vitamin D analogue and once daily coal tar polytherapy.

Results of the network meta-analysis indicate that there is a non-significant trend toward combined product containing calcipotriol monohydrate and betamethasone dipropionate being less effective than once and twice daily very potent corticosteroids in the treatment of patients with scalp psoriasis.

8.2.5. Cost effectiveness evidence (scalp psoriasis)

8.2.5.1. Economic evidence – literature review (scalp psoriasis)

One study6 was included that included relevant comparisons. It is summarised in the economic evidence profile below (Table 72 and Table 73). See also the full study evidence tables in Appendix I. No studies were excluded.

Table 72. Economic study characteristics.

Table 72

Economic study characteristics.

Table 73. Economic summary of findings.

Table 73

Economic summary of findings.

Although not presented in the above profile because they were dominated, it is worth noting themes from the overall analysis of all 12 treatment comparators. Overall, strategies that did not include combined or concurrent vitamin D or vitamin D analogue and potent corticosteroids (one applied in the morning and one in the evening) generated fewer QALYs and higher costs than those that did. In fact, the analysis showed that a strategy of starting with vitamin D or vitamin D analogue once daily and escalating to twice daily and then moving finally to Capasal (salicylic acid and coal tar shampoo) once daily was the most costly and the least effective of all 12 strategies.

There was little difference between the overall effectiveness (QALYs gained) of strategies depending upon when in the sequence the combined product containing calcipotriol monohydrate and betamethasone dipropionate came (first-, second- or third-line). Costs also did not seem to follow a pattern based on where combination product came in the sequence, but seemed to be driven more by what other treatments were in the sequence (e.g. once or twice daily vitamin D or vitamin D analogue and/or potent corticosteroid).

8.2.5.2. Economic evidence – original economic analysis (scalp psoriasis)

The review of clinical evidence for topical therapies used in the treatment of individuals with moderate to severe scalp psoriasis showed that there were several treatment options – tars, corticosteroids (potent and very potent), vitamin D or vitamin D analogues and combination products – each associated with certain advantages and disadvantages. The results of the network meta-analysis indicated that some interventions, such as very potent corticosteroid as well as combined product containing calcipotriol monohydrate and betamethasone dipropionate, were more likely to induce clearance or near clearance than others. Given that these combined and concurrent application strategies carry additional cost compared to both their individual constituent parts and compared to other topical alternatives, it was important to consider whether these additional costs are justified by additional health benefits in terms of improved quality of life.

The choice of which topical therapy to offer patients with moderate to severe scalp psoriasis in primary care was identified as among the highest economic priorities by the GDG because scalp psoriasis affects a large proportion of patients and is typically managed in primary care. As with topicals used to treat other body sites, even if the unit costs of the interventions are quite modest, the population affected is relatively large; therefore the health economic impact of any recommendation is likely to be substantial.

One cost-effectiveness analysis was identified in the published literature, but it had methodological limitations that called its conclusions into question. The analysis by Affleck6 did not include all of the relevant comparators under consideration for the guideline, namely very potent corticosteroids. Furthermore, the treatment effects used in their analysis differed from those found in the NCGC clinical review and network meta-analysis, and this difference was considered likely to affect the conclusion of the analysis. Due to these methodological limitations, there was still substantial uncertainty as to which topical therapy or therapies represented the best value for NHS resources in the treatment of scalp psoriasis. In order to reduce this uncertainty, an original cost-effectiveness analysis was undertaken by the guideline health economist in collaboration with the GDG. Below is a summary of the analysis that was undertaken. For full details please see Appendix N.

8.2.5.3. Methods

An analysis was undertaken to evaluate the relative cost-effectiveness of different topical therapy sequences used in the treatment of individuals with moderate to severe scalp psoriasis. A Markov model was used to estimate 12-month costs and quality-adjusted life years (QALYs) from a current UK NHS and personal social services perspective. A 12-month time horizon was considered clinically relevant and sufficiently long enough to capture important costs and consequences of first-line treatment in primary care. Uncertainty was explored through probabilistic analysis and sensitivity analysis. The performance of alternative treatment sequences was estimated using incremental cost-effectiveness ratios (ICERs), defined as the added cost of a given strategy divided by its added benefit compared with the next most expensive strategy. A threshold of £ 20,000 per QALY gained was used to assess cost-effectiveness.

The aim of the analysis was to identify the most cost-effective sequence of first, second and third line topical therapies for scalp psoriasis. It was important to model sequences given that most patients will commence treatment with one topical and then try others before moving on to more intensive treatments such as specialist applied topicals and/or systemic therapy. Table 74 presents the list of possible first, second and third line scalp treatments which may be combined in a sequence.

Table 74. Possible sequences of first, second and third line treatment.

Table 74

Possible sequences of first, second and third line treatment.

The following conditions were placed on the sequences, ensuring that they represented logical clinical practice:

  • Once daily treatment with a given topical would not come after a failure of twice daily treatment with the same topical;
  • Once daily treatment with potent steroid or vitamin D or vitamin D analogue would not come after once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate;
  • Once or twice daily treatment with potent corticosteroid would not come after once or twice daily with very potent corticosteroid.

Most comparators focus on evaluating a trial of three different treatments before referral for specialist review, but the GDG was also interested in whether earlier escalation of care might be more cost-effective. To test this, strategies have also been combined into two-treatment sequences with referral following a failure of second line treatment.

Due to the unacceptability of coal tar as a routine treatment (strong and unpleasant odours), this treatment was reserved for third line treatment only. This reflects their current placement in primary care given the availability of more acceptable and effective topicals such as those being compared as first and second line topicals.

The structure of the model developed by the NCGC was adapted from the model developed by Affleck and colleagues6 and was validated by the GDG as a reasonable reflection of current clinical practice. The Markov model and how patients move through the pathway is illustrated in Figure 7. Key model assumptions (these are discussed in more detail in the full write-up in Appendix N):

Figure 7. Patient flow diagram for the Markov model of topical treatments for scalp psoriasis.

Figure 7

Patient flow diagram for the Markov model of topical treatments for scalp psoriasis.

  • All hypothetical patients commence treatment with a given topical and experience one of two outcomes after 4 or 8 weeks:
    • response (defined as clearance/near clearance of their scalp psoriasis) or
    • no response (defined as something less than clearance/near clearance of their scalp psoriasis).
  • Patients who respond stop treatment and they either maintain response in the absence of treatment or they relapse.
    • Patients who relapse resume treatment with the same topical and again face a probability of responding or not responding.
  • Patients who do not respond to a given topical after 8 weeks of treatment are assumed to return to their GP and receive a prescription for an alternative topical therapy.
  • Patients can receive up to three different topical therapies before being referred by the GP to a specialist review in an outpatient dermatology clinic where second-line treatment options could be considered.
    • Some proportion of these referred patients will be kept on topical therapies, receive support and advice at the review consultation and be discharged back to their GP for long-term management.
    • Some will be treated by a specialist over 3 appointments in outpatient dermatology
    • The remaining proportion undergo a supervised scalp treatment with intensive topical therapy over the course of 3 dermatology day centre appointments:

      If they respond to intensive topical therapy they are then discharged to their GP for long-term management.

      If they do not respond to intensive topical therapy they continue to be managed by a specialist.

Movement between various health states is governed by transition probabilities, derived from the systematic review of clinical effectiveness data and network meta-analysis. Thirteen 4-week cycles were modelled, resulting in a 1-year time horizon for the analysis, with a half-cycle correction applied.

Model inputs were based on the clinical effectiveness review undertaken for the guideline, other published data and expert opinion where required. These are described in full in the technical report in Appendix N. All model inputs and assumptions were validated by the GDG.

8.2.5.4. Results

This analysis found that, given a NICE willingness-to-pay threshold of £ 20,000 per QALY gained, the most effective and cost-effective strategy is likely to be one of starting with once daily very potent corticosteroid and then escalating to twice daily very potent corticosteroid and then trying once daily TCF product if very potent steroids alone are insufficient to induce clearance or near clearance. This conclusion was based on the comparison of mean costs and mean QALYs across 169 modelled sequences. Base case results for non-dominated and non-extendedly dominated strategies are presented in Table 75. By starting with very potent corticosteroids once and then twice daily followed by TCF product was expected to generate 0.0014 more QALYs for an additional cost of £ 26.80 compared to the least costly sequence (once daily potent corticosteroid followed by once and then twice daily very potent corticosteroids). This gives and ICER of £ 19,143 per QALY gained, which is just under the NICE cost-effectiveness threshold. Based on total net monetary benefits and probabilities of being most cost-effective, there is little difference between the two strategies.

Table 75. Incremental analysis of base case results – scalp psoriasis.

Table 75

Incremental analysis of base case results – scalp psoriasis.

Complete results for all 169 comparators can be found in Appendix N. Overall, results of the analysis showed that the most effective (and cost-effective) strategies involved use of potent and very potent corticosteroids in at least two lines of treatment.

Results also showed that a strategy of using vehicle gel or emollient with no active agent only was the most costly and least effective strategy, largely driven by the cost of referrals and specialist management for non-responders. Similarly, a strategy of prescribing coal tar polytherapy for ongoing management was only slightly more effective than continued use of vehicle gel and cost the third most of any treatment sequence. Compared to strategies relying heavily on corticosteroids, strategies that included once or twice daily vitamin D analogue were unlikely to be cost-effective regardless of where they came in a treatment sequence. This finding is driven by their relatively low rank in terms of effectiveness and their relatively high acquisition cost relative to potent and very potent corticosteroids. Two compound formulation product, although third most effective in the network meta-analysis, was found to be cost-effective only as a third line intervention following very potent corticosteroids. Like vitamin D analogues, its high unit cost compared to other cheaper and effective topicals makes it unlikely to represent reasonable value for NHS resources.

The probabilistic analysis indicates that there is a great deal of uncertainty as to which sequence is optimal (i.e. most cost-effective). No single sequence was most cost-effective at a £20,000 per QALY willingness to pay threshold in more than 30% of simulations; however, looking across strategies indicates that those starting with once daily potent corticosteroid were optimal in 43% of simulations. In 33% of all simulations, following once daily potent with once or twice daily very potent corticosteroid was optimal. In another 44% of simulations, a sequence starting with either once or twice daily very potent corticosteroid was likely to be most cost-effective. The remaining 13% of simulations indicated that twice daily potent corticosteroids was an optimal first line strategy. These trends can also be seen by looking at the rank order of strategies in Table 36 of Appendix N, which shows that those starting with potent and very potent corticosteroids have the highest mean net benefits. These statistics indicate that we can be reasonably confident that starting with once daily potent or very potent corticosteroid is going to bring the greatest benefit for resources used, and that escalating to a twice daily very potent corticosteroid is likely to provide further benefit at reasonable extra cost.

A series of scenario analysis suggested that the conclusions from the base case are somewhat sensitive to changes in assumptions made.

Scenario analyses – restricted comparators

The base case analysis put a few conditions on the way topicals could be sequences (see Table 74 in section 8.2.5.3. These did not restrict how potent and very potent corticosteroids fit into treatment sequences. The GDG expressed concern that this lack of restrictions may not fully reflect the way these topicals are and should be used in general practice. They indicated that much more caution is and should be used when prescribing potent and very potent corticosteroids for both continuous and intermittent use. The GDG was also concerned that the analysis did not fully capture the safety risks associated with the use of these agents. In a stepwise fashion, various additional restrictions were placed on the use of these agents in each sequence.

Scenario 1: In the first scenario, all strategies involving potent or very potent corticosteroids (including two compound formulation product) in all three lines of treatment were removed. The results confirmed the findings of the base case results in which once daily very potent corticosteroid then twice daily very potent corticosteroid was found to be most cost-effective as first and second-line treatments. However, in this scenario no further steroid could be prescribed; therefore vitamin D analogue was found to be the most cost-effective third line treatment, applied either once or twice daily.

Scenario 2: In the second scenario, no sequence could include the consecutive use of potent or very potent corticosteroid, including as part of TCF product. The results again showed the likely cost-effectiveness of strategies including potent and very potent corticosteroids. Here, starting with once daily very potent corticosteroids and then moving to once or twice daily vitamin D analogue and then twice daily very potent corticosteroids was least costly and second most effective. Starting the sequence with twice daily very potent corticosteroid and ending with once daily TCF product generated 0.00055 more QALYs, but at an additional cost of £45.20 per year. The resulting ICER (£82,182) is thus over the £20,000 per QALY threshold.

Scenario 3: In the third scenario, twice daily application of very potent corticosteroid could not precede once daily application. There were no changes to the base case results under these conditions.

Scenario 4: If the conditions outlined in scenarios 1 and 2 are combined and very potent corticosteroids were also restricted such that they could not appear first in a sequence, then the optimal strategy at a £20,000 per QALY threshold is to start with once daily potent corticosteroid, then move to twice daily vitamin D and end with once or twice daily very potent corticosteroid. Replacing first line potent steroid with once daily TCF product is expected to generate <0.0007 QALYs, but for an additional cost of around £145 (ICER>£200,000).

In addition to the concerns raised about the safety of potent and very potent corticosteroids, the GDG raised the issue of cosmetic acceptability and its importance in the treatment of scalp psoriasis. In particular, they voiced a strong preference for once daily application, stating that few patients would be willing or interested in applying topicals to their scalp more than once a day, at night. On that basis, modelled comparators were restricted in a stepwise fashion.

Scenario 5: In the fifth scenario, twice daily strategies were reserved for second and third line treatment following failure of at least one once daily strategy. Under this scenario and combined with the restrictions outlined in scenario 4 above, the optimal sequence was once daily potent corticosteroids followed by once or twice daily vitamin D, and ending with once or twice daily very potent corticosteroid.

Replacing initial potent corticosteroids with once daily TCF product in this sequence would increase benefits (0.00058 QALYs) but also increase cost (£147) at a ratio of £253,621 per QALY gained. Similarly, replacing second line vitamin D analogue with once daily TCF product would produce additional QALY gains (approximately 0.001), but at extra cost (approximately £40), producing ICERs around £40,000 per QALY gained. Scenario 6: In a final scenario, all twice daily strategies were removed and only sequences of once daily treatments were included. If steroids could be offered anywhere in the sequence, then the most cost-effective strategy was to start with potent corticosteroids, move up to very potent corticosteroids and then try TCF product if both steroids alone have failed. If one wishes to avoid consecutive use of steroids, then the optimal strategy is to start with potent steroids, then switch to vitamin D analogues and end with very potent corticosteroids. Replacing very potent corticosteroids with TCF product in this sequence generates 0.00132 more QALYs, but with an ICER too high to be considered cost-effective (ICER=£39,773).

Sensitivity analyses – Variation in early versus late response

The base case assumed that patients would trial a given topical for up to 8 weeks (maximum 4 weeks for very potent corticosteroids). Some proportion would be expected to respond by 4 weeks, and discontinue treatment at that time. The remainder would carry on to 8 weeks, at which time non-responders would move on to the next topical in a sequence. The data defining the breakdown of early (at 4 weeks) vs late (at 8 weeks) responders came from three studies169,199,407 and was thus uncertain. Deterministic sensitivity analyses were performed around these parameters to observe the impact on the results.

First, an analysis was performed in which no one was expected to respond and discontinue treatment at 4 weeks (i.e. all responders require 8 weeks treatment). Compared to the results of the base case when all comparators are included, the ICER for once and then twice daily very potent corticosteroids followed by once daily TCF product increased to over £20,000 per QALY, making once daily potent corticosteroids followed by once and then twice daily very potent corticosteroids the optimal sequence. No changes to the conclusions of the more restrictive scenario 5 were observed (i.e. once daily potent corticosteroids then once or twice daily vitamin D followed by once or twice daily very potent corticosteroid is still optimal).

Second, an analysis was performed in which all responders were assumed to respond by 4 weeks, with no one requiring an additional 4 weeks of treatment. Small reductions in total cost and small improvements in total benefits were observed, but no significant changes to the results of the base case were observed.

Finally, an analysis was performed in which a 4-week stopping rule was applied. In this scenario, responders were limited to those that have responded by week 4 (see Appendix N), and all other patients are assumed to move on to the next topical in the sequence (i.e. no one continues to 8 weeks of treatment with the same topical). The results of the base case were only somewhat sensitive to this stopping rule, with total costs and benefits improving slightly. Third line TCF product after once and twice daily very potent corticosteroids became even more cost-effective than in the base case. In the context of scenario 5, however, third line TCF product instead of once or twice daily very potent corticosteroids is still too costly relative to its added benefit to represent good value for NHS resource given the NICE threshold of £20,000.

Sensitivity analyses – Reduced adherence

There was some concern that issues of treatment adherence were inadequately captured in the model. The estimates of effect used in the base case were derived from randomised controlled trials which may represent the best case scenario for topical therapies. The GDG wished to explore how reduced adherence to twice daily treatments would affect the conclusions of the base case. In this scenario, 60% of patients being treated with twice daily topical were assumed to adhere to treatment whilst the remaining 40% of patients were assumed to apply the topical only once daily. Thus, efficacy of the treatment would be reduced compared to the base case estimates. To be conservative, no reductions in cost were assumed despite the fact that less topical would be used. With adherence reduced, the optimal strategy when all 169 comparators were included was once daily potent corticosteroid followed by once and then twice daily very potent corticosteroid. This was the second most cost-effective strategy in the base case. When considering only strategies included in Scenario 5 above, conclusions do not change. Once daily potent corticosteroid followed by once or twice daily vitamin D and then once or twice daily very potent corticosteroids is still optimal at a £20,000 threshold.

Sensitivity analysis - Lower expected resource use for combined product containing calcipotriol monohydrate and betamethasone dipropionate

The base case of this analysis assumed that patients using combined product containing calcipotriol monohydrate and betamethasone dipropionate for 4 weeks would use approximate 71.4 g of product. This estimate was based on the mean across five RCTs48,168,169,405,408. In a recent UK cost-utility analysis, Affleck and colleagues6 assumed the 4-week quantity used to be 60 g. At this quantity, the unit cost of combined product containing calcipotriol monohydrate and betamethasone dipropionate is cut nearly in half. This value was used in a sensitivity analysis to explore how sensitivity the results were to this particular value. This was quite a favourable scenario for TCF product as costs were reduced without assuming any commiserate reduction in efficacy by using less topical.

The results suggest that the base case conclusions, for which all sequences are included, do not change when the dose of TCF is fixed at 60 g. Here, as in the base case, the most effective and cost-effective strategy places once daily TCF product as a third line treatment after trials of once and then twice daily very potent corticosteroid. The ICER comes down to under £1,000 in this sensitivity analysis compared to just over £19,000 in the base case.

Conclusions from the various scenarios in which most comparators are removed from the analysis for reasons of safety and patient preference (Scenario 5), appear to be somewhat sensitive to reductions in assumed dose of TCF product.

First line use of TCF product is still unlikely to represent better value for NHS resources than potent corticosteroids alone. To replace once daily potent corticosteroids with once daily TCF product as first line in a sequence followed by once or twice daily vitamin D analogue and then once or twice daily very potent corticosteroids would cost more than £70,000 per additional QALY gained. Although this is lower than the ICERs when base case dosing assumptions are in effect (ICERs >£180,000), it is still not low enough to be considered cost-effective given the NICE willingness to pay threshold.

Under base case dosing assumptions, as a second line strategy after once daily potent corticosteroid once daily TCF product was unlikely to be cost-effective compared to second line once and twice daily vitamin D (ICERs >£30,000 per QALY). When usage is assumed not to exceed 60 g per 4 weeks, then second line once daily TCF product is likely to dominate (be less costly and more effective than) once and twice daily vitamin D.

Finally, when only once daily treatments are considered, as in scenario 6 above, reduced 4-week usage of TCF product brings the ICER of third line TCF product compared to very potent corticosteroid (following potent steroid and vitamin D) down to £5,279 compared to £39,733.

Sensitivity analyses – unit cost of potent corticosteroids

The base case assumed that the cost for each topical was based on the product and scalp formulation with the lowest unit cost per gram/millilitre. Given that clinicians and patients may have preferences for different products or formulations, it was considered necessary to explore how variation in price of topicals, particularly potent corticosteroids, might affect the results. To do this, the highest cost (per gram) potent corticosteroid Synalar gel (fluocinolone acetonide) was assumed in place of Betacap scalp application. The cost of Synalar gel is around 4.6 times that of Betacap scalp application.

Under this costing assumption and considering all comparators, the sequence of once then twice daily very potent corticosteroid followed by once daily TCF product becomes the most effective and least costly. It is now less costly than the strategy starting with potent corticosteroids and then escalating up to once then twice daily very potent corticosteroids.

Additionally, the results of scenario 5, in which twice daily treatments and very potent corticosteroids are reserved for second and third line treatment and corticosteroids cannot be used consecutively, were insensitive to increased costs. The strategy of starting with once daily potent corticosteroid followed by once or twice vitamin D and then finally once or twice daily very potent corticosteroid remains the optimal choice given a £20,000 per QALY threshold.

Sensitivity analyses – model time horizon

A one year time horizon was used in the base case on the basis that little is known about the longer term efficacy, adherence and course of moderate to severe scalp psoriasis. Aware the psoriasis, including scalp psoriasis, is a chronic and long term condition, the GDG chose to explore how the results might be affected by lengthening the model time horizon to 2, 3 and 5 years. The results of the base case, where all 169 comparators are included, appear somewhat sensitive to changes in the time horizon. The most effective and cost-effective strategy in the base case (once and then twice daily very potent corticosteroid followed by once daily TCF product) is still most effective at 2, 3 and 5 years; however, its ICER relative to the least cost and second most effective sequence (once daily potent corticosteroid followed by once and then twice daily very potent corticosteroid) increases to values over the £20,000 threshold (£39,000, £56,000 and £73,000 at 2, 3 and 5 years respectively).

The results of scenarios 5 and 6 (as outlined above), wherein comparators are restricted in certain ways, are insensitive to extensions of the time horizon. Once daily potent corticosteroid followed by once or twice daily vitamin D and then once or twice daily very potent corticosteroid are still optimal.

8.2.5.5. Interpretation and limitations

In assessing the relative cost-effectiveness of alternative topical therapies in patients with moderate to severe scalp psoriasis limited evidence was available from the published economic literature. The evidence that was identified and included in the health economic review had potentially serious limitations and therefore the GDG considered it a priority to undertake original evaluation for the guideline in order to inform recommendations.

Original decision modelling undertaken for the guideline showed that there were relatively small differences in terms of benefit between 169 different topical sequences, but the differences in terms of cost were quite substantial. Based on the mean costs and benefits, the analysis suggests that initial treatment with once daily very potent corticosteroid followed by twice daily very potent corticosteroid and then once daily TCF product if very potent corticosteroids alone are insufficient to induce clearance or near clearance is likely to represent the most cost-effective sequence for moderate to severe scalp psoriasis. Uncertainties in the analysis were explored through sensitivity analysis which showed that in some scenarios in which restrictions were placed on the comparators

  • Once daily potent corticosteroid is likely to be the optimal first line treatment if very potent corticosteroids are considered too aggressive.
  • Once or twice daily vitamin D or analogues are likely to be cost-effective second in the sequence, after trials of potent or very potent corticosteroids, particularly where continuous corticosteroids are to be avoided
  • Once or twice daily very potent corticosteroids is likely to be the most cost-effective third line treatment if potent corticosteroid and vitamin D have not worked
  • TCF product may be cost-effective, but only after potent and/or very potent corticosteroids have failed and when only once daily applications of topicals is being considered

In general, sequences including once daily TCF product were slightly more effective than the same sequence including alternatives such as vitamin D analogue or potent corticosteroid; however, the very modest additional benefits (<0.001 and dependent on comparator) would only be considered potentially cost-effective if willingness to pay thresholds were substantially greater than £20,000 per QALY gained. If, however, the amount of TCF product used by patients is less than reported in the clinical trial evidence, such that a single 60 g pack is needed for 4 weeks, then TCF product may be cost-effective as a second or third line treatment following potent corticosteroids. Under no conditions was first line use of TCF product likely to represent better value for NHS resources than potent or very potent corticosteroids.

The analysis has several limitations which were considered carefully by the GDG. Firstly, the analysis evaluates treatment sequences even though the available trial data compares single topicals head to head without sequencing. In order to apply the treatment effects within the sequencing model, we assumed that treatment effects were independent. That is, we assumed the effectiveness of the combined product containing calcipotriol monohydrate and betamethasone dipropionate as a second or third line topical was equal to its effectiveness as a first line agent and that this was true regardless of other topicals it may follow. The GDG did not believe this to be a significant limitation given that the patients included in the overwhelming majority of RCTs were reported to have psoriasis for longer than 5 years, during which they can be assumed to have previously tried, succeeded and/or failed various topical treatments.

The analysis only captured the efficacy of topicals and did not capture the costs or consequences of adverse events. Although the RCT evidence on adverse events was sparse, the GDG is conscious of the risks associated with the long-term use of potent and very potent corticosteroids. They carefully considered whether the added effect in terms of clearance was worth the potential risks of adverse effects.

The model was also focused on the induction of disease clearance as opposed to the maintenance of clearance. No trials focusing on maintenance were identified in the clinical evidence review and therefore no evidence was available for use in the economic model.

The model also takes a relatively short time horizon considering that psoriasis of the scalp is a chronic, long term condition for which patients may take up treatment intermittently for many years of their lives. Frequency and severity of relapse, selection for and speed of onward referral, methods of self-management and long-term safety are all issues inadequately addressed in the evidence base and therefore translate into limitations of the economic analysis. Longer time horizons of up to 5 years were explored in sensitivity analyses and conclusions were insensitive to these extensions.

The model estimated the health gain for each treatment by mapping the change in PASI score to the EQ-5D based on observational evidence. However, it has been noted that several important areas of health-related quality of life for people with psoriasis are not directly assessed by the EQ-5D questionnaire226. Therefore it is possible that the EQ-5D may lack content validity for these patients. Research is ongoing in this area. But we note that even using a £30,000 per QALY threshold rather than £20,000 would not change the conclusions of our analyses. Therefore only if the EQ-5D is under-estimating health gain of one treatment compared to another by a considerable extent, could this pose a serious limitation.

This analysis of the treatment of psoriasis of the scalp is distinct from the analysis of the treatment of scalp of the trunk and/or limbs largely because it is based on a different evidence base and as such has given rise to site-specific recommendations. In clinical practice, healthcare professionals are likely to see patients who are dealing with psoriasis at a variety of sites, including their face and flexures. It is quite possible that healthcare professionals will need to prescribe different topicals for different sites, meaning that patients may have several different agents at a time. Indeed, even if they are using the same product (i.e. potent corticosteroid) on different sites, they may be prescribed different formulations for each site (i.e. creams or ointments for the trunk and limbs; gels or foams for the scalp). It would be simpler to prescribe one single treatment for all sites, but as the clinical and cost-effectiveness has shown, such an approach may not represent the most effective or efficient use of NHS resources.

8.2.5.6. Comparison with published studies

The findings from the NCGC original economic analysis are quite different from the results of the most similar published study by Affleck and colleagues6. Affleck and colleagues found a sequence starting with twice daily potent corticosteroids followed by concurrent treatment with vitamin D or vitamin D analogue and potent corticosteroid corticosteroids (one applied in the morning and one in the evening) and then once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate to be most cost-effective. Although the analysis appears to have been executed well, the included comparators and the estimates of effect and resource use had limitations which called the conclusions of the analysis into question.

The biggest differences in the results of the NCGC analysis presented here and the analysis undertaken by Affleck has to do with the comparators included, namely the inclusion/exclusion of very potent corticosteroids. The NCGC analysis included very potent corticosteroids as the network meta-analysis demonstrated them to be highly efficacious in the short term treatment of psoriasis of the scalp. The GDG confirmed that although very potent corticosteroids are not normal management for the treatment of the trunks and limbs, they constitute a reasonable, short-term option for treating the scalp.

The second key difference between the analyses relates to the relative treatment effects used. Affleck and colleagues derived their treatment effects from an adjusted indirect comparison38, which, when compared to the NCGC network meta-analysis, appears to have overestimated the effectiveness of TCF product compared to other topicals. For example, in their analysis TCF product was found to be 2.45 times more likely to induce response than once daily calcipotriol (RR=2.45, 95% CI: 1.84 to 3.27). The NCGC network meta-analysis found the risk ratio to be lower, around 1.857. This translates into an absolute risk difference between the two comparators of 35.54% using Affleck’s estimates and 29.65% using the NCGC estimates. Differences such as these add up when synthesised in economic models and could lead to biased conclusions.

In addition, the estimate they used for quantity of TCF product used per 4-week treatment period was 60 g, compared to the estimate used in the NCGC analysis 71.4 g. Based on these estimates of resource use, the NCGC analysis assumes 4 weeks of TCF product costs £31.29 more than Affleck and colleagues did. We performed a sensitivity analysis in which we assumed the same quantity of TCF product used by Affleck and colleagues (i.e. 60 g, £36.50). The ICER for TCF product as a third line treatment improved significantly compared to the base case, making it potentially cost-effective given the NICE willingness to pay threshold. However, there remains a great deal of uncertainty in this conclusion.

One thing that Affleck and colleagues were able to capture that the NCGC analysis was not had to do with the potential disutilities associated with adverse events. They included these in their base case, and unfortunately did not report a sensitivity analysis wherein they were removed altogether with which to compare. However, the authors did state that variation in the incidence of adverse events, upwards and downwards, did not change the conclusions of their analysis.

8.2.5.7. Evidence statements

  • One directly applicable study with potentially serious limitations found that a sequence of potent corticosteroid followed by concurrent vitamin D or vitamin D analogue and potent corticosteroid corticosteroids (one applied in the morning and one in the evening) and followed by the combined product containing calcipotriol monohydrate and betamethasone dipropionate to be the most cost-effective strategy to treat chronic scalp psoriasis.
  • One directly applicable study with potentially serious limitations found that treatment sequences that do not include combined or concurrent vitamin D or vitamin D analogue and potent corticosteroids (one applied in the morning and one in the evening) are among the least effective and most costly in the treatment of chronic scalp psoriasis.
  • New economic analysis from a current UK NHS and PSS perspective comparing 169 different sequences of topical therapies found sequences beginning with once daily very potent corticosteroids to offer the best value for NHS resource in the treatment of patients with moderate to severe scalp psoriasis; however, this conclusion was sensitive to many sensitivity and scenario analyses undertaken.
    • The most consistently cost-effective first line treatment when very potent corticosteroids were excluded was once daily potent corticosteroid. This conclusion was robust to the majority of sensitivity and scenario analyses undertaken.
    • Choice of second and third line treatments was more uncertain, but very potent corticosteroids, once or twice daily, were generally shown to be most cost effective, followed in rank order by once or twice daily vitamin D or analogue and then once daily two-compound formulation product. This conclusion was somewhat sensitive to alternative assumptions regarding suitability and acceptability of certain comparators.

      Sensitivity analyses in which continuous or consecutive use of topicals containing steroids was restricted found that once and twice daily vitamin D analogues are cost-effective as second line treatments in sequences with potent and very potent corticosteroids.

      Sensitivity analyses in which only once daily applications were considered found that initial treatment with potent steroids was optimal, followed by either very potent corticosteroid and then two-compound formulation product if steroids could be used continuously or followed by vitamin D analogue and very potent corticosteroid if continued use of steroids was to be avoided.

8.2.6. Face, flexures and genitals

There were 3 studies that addressed the efficacy and safety of topical treatments for psoriasis affecting the face and/or flexures (including genitals).

  • One study215 combined people treated for affected skin on the face and intertriginous areas (proportions not given)
  • One study129 included only inverse/flexural sites
  • One study220 combined people treated for affected skin on the face and genitofemoral areas (90% had lesions on the face and 10% on the genitofemoral sites)

8.2.6.1. Tacrolimus vs. placebo

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsTacrolimusPlaceboRelative (95% CI)Absolute
Investigator's assessment (clear/nearly clear) – Tacrolimus BD (follow-up 8 weeks)
1
Lebwohl 2004
randomised trialsvery seriousano serious inconsistencyno serious indirectnessno serious imprecisionnone73/112 (65.2%)17/55 (30.9%)RR 2.11 (1.39 to 3.2)343 more per 1000 (from 121 more to 680 more)⊕⊕○○
LOW
Withdrawals due to adverse events – Tacrolimus BD (follow-up 8 weeks)
1
Lebwohl 2004
randomised trialsvery seriousano serious inconsistencyno serious indirectnessvery seriousbThe adverse event was not at the treatment site0/98 (0%)1/40 (2.5%)RR 0.14 (0.01 to 3.32)22 fewer per 1000 (from 25 fewer to 58 more)⊕○○○
VERY LOW
Withdrawals due to lack of efficacy – Tacrolimus BD (follow-up 8 weeks)
1
Lebwohl 2004
randomised trialsvery seriousano serious inconsistencyno serious indirectnessno serious imprecisionnone0/98 (0%)6/45 (13.3%)RR 0.04 (0 to 0.62)128 fewer per 1000 (from 51 fewer to 133 fewer)⊕⊕○○
LOW
a

Unclear allocation concealment and blinding; high dropout rate in placebo group (29.1% vs 12.5% in tacrolimus group)

b

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

Evidence statements

In people with chronic plaque psoriasis affecting the face and/or intertriginous areas, tacrolimus twice daily was statistically significantly better than placebo for:

  • Investigator’s assessment (clear/nearly clear) at 8 weeks [1 study; 167 participants; low quality evidence]215
  • Withdrawal due to lack of efficacy at 8 weeks [1 study; 143 participants; low quality evidence]215

In people with chronic plaque psoriasis affecting the face and/or intertriginous areas, there was no statistically significantly difference between tacrolimus twice daily and placebo for:

  • Withdrawal due to adverse events at 8 weeks [1 study; 138 participants; very low quality evidence]215

8.2.6.2. Pimecrolimus vs. placebo

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsPimecrolimusPlaceboRelative (95% CI)Absolute
Investigator's assessment (clear/nearly clear) – Pimecrolimus BD (follow-up 8 weeks)
1
Gribetz 2004
randomised trialsno serious risk of biasano serious inconsistencyno serious indirectnessno serious imprecisionnone20/28 (71.4%)6/29 (20.7%)RR 3.45 (1.63 to 7.31)507 more per 1000 (from 130 more to 1000 more)⊕⊕⊕⊕
HIGH
Withdrawals due to adverse events – Pimecrolimus BD (follow-up 8 weeks)
1
Gribetz 2004
randomised trialsno serious risk of biasano serious inconsistencyno serious indirectnessno serious imprecisionnone0/26 (0%)0/25 (0%)not poolednot pooled⊕⊕⊕⊕
HIGH
Withdrawals due to lack of efficacy – Pimecrolimus BD (follow-up 8 weeks)
1
Gribetz 2004
randomised trialsno serious risk of biasano serious inconsistencyno serious indirectnessvery seriousbnone1/27 (3.7%)2/27 (7.4%)RR 0.50 (0.05 to 5.19)37 fewer per 1000 (from 70 fewer to 310 more)⊕⊕○○
LOW
Skin atrophy – Pimecrolimus BD (follow-up 8 weeks)
1
Gribetz 2004
randomised trialsno serious risk of biasano serious inconsistencyno serious indirectnessno serious imprecisionnone0/28 (0%)0/29 (0%)not poolednot pooled⊕⊕⊕⊕
HIGH
a

Higher drop-out in placebo group (13.8% vs 7.1% in pimecrolimus group) but rates acceptable in both groups

b

Confidence interval crosses the boundary for clinical significance in favour of both treatments, as well as line of no effect

Evidence statements

In people with chronic plaque psoriasis affecting the flexural areas, pimecrolimus twice daily was statistically significantly better than placebo for:

  • Investigator’s assessment (clear/nearly clear) at 8 weeks [1 study; 57 participants; high quality evidence]129

In people with chronic plaque psoriasis affecting the flexural areas, there were no events with either pimecrolimus twice daily or placebo for:

  • Withdrawal due to adverse events at 8 weeks [1 study; 51 participants; high quality evidence]129
  • Skin atrophy at 8 weeks [1 study; 57 participants; high quality evidence]129

In people with chronic plaque psoriasis affecting the flexural areas, there was no statistically significant difference between pimecrolimus twice daily and placebo for:

  • Withdrawal due to lack of efficacy at 8 weeks [1 study; 54 participants; low quality evidence]129

8.2.6.3. Tacrolimus vs. vitamin D or vitamin D analogue

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsTacrolimusVitamin D or vitamin D analogueRelative (95% CI)Absolute
Investigator's assessment (clear/nearly clear) – Tacrolimus BD vs calcitriol BD (follow-up 6 weeks)
1
Liao 2007
randomised trialsseriousano serious inconsistencyno serious indirectnessseriousbnone15/25 (60%)8/24 (33.3%)RR 1.8 (0.94 to 3.45)267 more per 1000 (from 20 fewer to 817 more)⊕⊕○○
LOW
Withdrawals due to adverse events – Tacrolimus BD vs calcitriol BD (follow-up 6 weeks)
1
Liao 2007
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone0/25 (0%)0/21 (0%)not poolednot pooled⊕⊕⊕○
MODERATE
Withdrawals due to lack of efficacy – Tacrolimus BD vs calcitriol BD (follow-up 6 weeks)
1
Liao 2007
randomised trialsseriousano serious inconsistencyno serious indirectnessno serious imprecisionnone0/25 (0%)0/21 (0%)not poolednot pooled⊕⊕⊕○
MODERATE
a

Unclear allocation concealment and not matched at baseline for sex or disease severity (less severe and fewer men in tacrolimus group); higher dropout rate in calcipotriol group (12% vs 0% on tacrolimus)

b

Confidence interval ranges from clinically important effect to no effect

In people with chronic plaque psoriasis affecting the face and/or genitofemoral areas, there were no events with either tacrolimus twice daily or vitamin D (calcitriol twice daily) for:

  • Withdrawal due to adverse events at 6 weeks [1 study; 46 participants; moderate quality evidence]220
  • Withdrawal due lack of efficacy at 6 weeks [1 study; 46 participants; moderate quality evidence]220

In people with chronic plaque psoriasis affecting the face and/or genitofemoral areas, there was no statistically significant difference between tacrolimus twice daily and vitamin D (calcitriol twice daily) for:

  • Investigator’s assessment (clear/nearly clear) at 6 weeks [1 study; 49 participants; low quality evidence]220

8.2.7. Time to remission or maximum effect for face, flexures and genitals

8.2.7.1. Tacrolimus

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsTacrolimus BD
Mean time to maximum response (PGA) (follow-up 57 days)
1
Lebwohl 2004
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessno serious imprecisionnoneTacrolimus 0.1%

112
Patients achieving excellent improvement or clearing

Day 8: 24.8%

Day 57: 66.7%
⊕⊕○○
LOW
Mean time to maximum response (PGA) (follow-up 57 days)
1
Lebwohl 2004
observational studiesano serious risk of biasbno serious inconsistencyno serious indirectnessvery seriouscnoneTacrolimus 0.1%

112
Based on graphical representation of the % with excellent improvement or clearing the majority of those who achieved success did so by day 29, with a small decrease in % to day 43 but a further increase of <5% between days 29 and 57⊕○○○
VERY LOW
Mean time to maximum response (PGA) (follow-up 6 weeks)
1
Liao 2007
observational studiesano serious risk of biasbno serious inconsistencyseriousdvery seriouscnoneTacrolimus 0.03%

25
Graphical representation of % clear or nearly clear over time demonstrated that maximum effect was reached not reached by week 6⊕○○○
VERY LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

c

Interpreted from graphical representation

d

Incorrect outcome measure

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for topical tacrolimus (no statistical analysis could be performed).

In people with face/flexural psoriasis, the time to remission when using tacrolimus varied between studies:

  • Proportion achieving remission on tacrolimus 0.1% by 57 days was 66.7% [1 study; 112 participants; low quality evidence]215
  • Of those who achieved remission on tacrolimus 0.1% by the end of the trial, 37.2% had responded by day 8 based on investigators assessment [1 study; 112 participants; low quality evidence]215
  • Mean time to remission on tacrolimus 0.1% on PGA showed that a maximum effect was reached by week 4 [1 study; 112 participants; very low quality evidence]215
  • Mean time to maximum response based on tacrolimus 0.03% on PGA showed that a maximum effect was not reached by week 4 [1 study; 25 participants; very low quality evidence]220
Summary

The evidence suggests that maximum response to tacrolimus 0.1% is achieved by 4 weeks of treatment, but maximum response is later when using a lower concentration215,220.

8.2.7.2. Pimecrolimus

Evidence profile
Quality assessmentNo of patientsEffectQuality
No of studiesDesignRisk of biasInconsistencyIndirectnessImprecisionOther considerationsPimecrolimus 1% BD
Time-to-clear/nearly clear (follow-up 8 weeks)
1
Gribetz 2004
observational studies1no serious risk of bias2no serious inconsistencyno serious indirectnessno serious imprecisionnone28Percentage of patients clear or almost clear

Baseline: 0%

Day 3: 14.3%

Day 7: 35.7%

Week 2: 53.6%

Week 4: 64.3%

Week 6: 67.9%

Week 8: 71.4%
⊕⊕○○
LOW
a

Although the data are taken from randomised trials the benefit of control data is not being utilised as considerations are being made based on single interventions without reference to the comparator arm

b

Unclear allocation concealment may have biased patient selection for this intervention

Evidence statements

Evidence statements for individual studies that provide data regarding the time to remission or time to maximum response for topical pimecrolimus (no statistical analysis could be performed).

In people with flexural psoriasis, the time to remission when using pimecrolimus was as follows:

  • Proportion achieving remission by 8 weeks was 71.4% [1 study; 28 participants; low quality evidence]129
  • The continued increase in responders between 6 and 8 weeks was 3.5% [1 study; 28 participants; low quality evidence]129
  • Some people (35.7%) achieved remission by 1 week [1 study; 28 participants; low quality evidence]129
  • Of those who achieved remission by the end of the trial (8 weeks), 75.1% had responded by week 2, 90.1% by week 4 and 95.1% by week 6 based on investigators assessment [1 study; 28 participants; low quality evidence]129

8.2.7.3. Summary

The evidence suggests that maximum response may be achieved by 8 weeks, with the continued response rate increasing only slightly between weeks 6 and 8129. However, the majority of those who will respond within 8 weeks had done so by week 4.

8.2.8. Cost effectiveness evidence – face and flexures (including genitals)

No relevant studies were identified. In the absence of recent UK cost-effectiveness analysis, relevant unit costs were sourced to aid consideration of cost effectiveness (Table 76).

Table 76. Costs of medications for face and flexures (including genitals).

Table 76

Costs of medications for face and flexures (including genitals).

8.2.9. Recommendations and link to evidence

Recommendations on topical treatment for scalp psoriasisTopical treatment of psoriasis affecting the scalp
48.

Offer a potent corticosteroidddd applied once daily for up to 4 weekseee as initial treatment for people with scalp psoriasis.

49.

Show people with scalp psoriasis (and their families or carers where appropriate) how to safely apply corticosteroid topical treatment.

50.

If treatment with a potent corticosteroidfff does not result in clearance, near clearance or satisfactory control of scalp psoriasis after 4 weeksggg consider:

  • a different formulation of the potent corticosteroid (for example, a shampoo or mousse) and/or
  • topical agents to remove adherent scale (for example, agents containing salicylic acid, emollients and oils) before application of the potent corticosteroid.
51.

If the response to treatment with a potent corticosteroidfff for scalp psoriasis remains unsatisfactory after a further 4 weeksggg,hhh of treatment offer:

  • a combined product containing calcipotriol monohydrate and betamethasone dipropionateiii applied once daily for up to 4 weeks or
  • vitamin D or a vitamin D analoguejjj applied once daily (only in those who cannot use steroids and with mild to moderate scalp psoriasis).
52.

If continuous treatment with either a combined product containing calcipotriol monohydrate and betamethasone dipropionateiii applied once daily or vitamin D or a vitamin D analogue applied once daily for up to 8 weeksggg does not result in clearance, near clearance or satisfactory control of scalp psoriasis offer:

  • a very potent corticosteroid applied up to twice daily for 2 weeks for adults only or
  • coal tar applied once or twice daily or
  • referral to a specialist for additional support with topical applications and/or advice on other treatment options.
53.

Consider topical vitamin D or a vitamin D analoguejjj,kkk alone for the treatment of scalp psoriasis only in people who:

  • are intolerant of or cannot use topical corticosteroids at this site or
  • have mild to moderate scalp psoriasis.
54.

Do not offer coal tar-based shampoos alone for the treatment of severe scalp psoriasis.

Recommendations on topical treatment for psoriasis of the face, flexures and genitalsTopical treatment of psoriasis affecting the face, flexures and genitals
55.

Offer a short-term mild or moderate potency corticosteroidlll applied once or twice daily (for a maximum of 2 weeksmmm) to people with psoriasis of the face, flexures or genitals.

56.

Be aware that the face, flexures and genitals are particularly vulnerable to steroid atrophy and that corticosteroids should only be used for short-term treatment of psoriasis (1–2 weeks per month). Explain the risks to people undergoing this treatment (and their families or carers where appropriate) and how to minimise them.

57.

For adults with psoriasis of the face, flexures or genitals if the response to short-term moderate potency corticosteroids is unsatisfactory, or they require continuous treatment to maintain control and there is serious risk of local corticosteroid-induced side effects, offer a calcineurin inhibitornnn applied twice daily for up to 4 weeks. Calcineurin inhibitors should be initiated by healthcare professionals with expertise in treating psoriasis.

58.

Do not use potent or very potent corticosteroids on the face, flexures or genitals.

59.

When prescribing topical agents at facial, flexural and genital sites take into account that they may cause irritation and inform people undergoing treatment (and their families and carers where appropriate) of these risks and how to minimise them. See also recommendation 56.

Future research recommendationsNone.
Relative values of different outcomesThe relative values of the different outcomes for scalp, face and flexural (including genital) sites are the same as for trunk and limbs.
  • Clear/nearly clear (investigator)
  • Clear/nearly clear (patient)
  • % change in PASI
  • Duration of remission
  • Withdrawal due to toxicity
  • Withdrawal due to lack of efficacy
  • Skin atrophy.
Based on the results from the pairwise and network meta-analyses and the health economic model the GDG recommended potent corticosteroids as the first topical intervention, followed by very potent steroids if this failed, as this was the most cost-effective option based on the investigator and patient assessment of achieving clear or nearly clear status. There was no clinically significant difference between most interventions in terms of withdrawal due to toxicity and skin atrophy, as the absolute numbers were low and clear evidence regarding duration of remission was lacking.

It was also noted that the pair-wise comparison of the combined product containing calcipotriol monohydrate and betamethasone dipropionate compared to potent steroid alone (applied once-daily for the scalp) did not show a clinically significant difference in efficacy. Unlike the comparison for treatment of the trunk and/or limbs.
Trade off between clinical benefits and harmsAs with the use of corticosteroids on the trunk and limbs, the efficacy, time to clearance and cosmetic acceptability were felt to outweigh the potential risks of corticosteroids for treatment of the scalp. The GDG discussed the data showing that of those who respond by 8 weeks to potent corticosteroid treatment, approximately 84% had done so by 4 weeks. Therefore, it was agreed to consider different formulations and topical agents to remove scale if treatment had not been successful by 4 weeks.

The GDG noted that, unlike at the trunk and limbs, from the scalp data there was no consistent trend linking frequency of application to improved efficacy. Once and twice daily vitamin D analogues were roughly equal in effect, whereas once daily potent corticosteroids may be better than twice daily and twice daily very potent corticosteroids may be better than once daily. The GDG thought that this may be a function of adherence and/or acceptability of twice daily scalp treatments, which are not generally favoured by patients. Their experience suggests that patients strongly prefer once daily scalp applications due to the messiness, inconvenience and cosmetic unacceptability of multiple applications each day. Therefore, to optimise outcomes once daily application was recommended where possible as well as emphasising the importance of using the correct formulation and removal of adherent scale, which is particularly important when treating scalp psoriasis.

When considering clinically appropriate sequences of treatment for scalp psoriasis, the GDG agreed that starting with a very potent corticosteroid as the first topical intervention would be an inappropriately aggressive strategy.

The GDG were more cautious when considering this trade off in favour of corticosteroids at face and flexural sites as risks of skin atrophy are higher. The GDG considered that only mild, or if necessary moderate potency corticosteroid could be justified. Calcineurin inhibitors, whilst effective, are unlicensed for psoriasis. The GDG considered that given the paucity of other options, the impact psoriasis has on these sites and also that these agents are licensed and widely used in eczema, calcineurin inhibitors could be recommended following specialist advice. Twice daily use is specified as this was the frequency of treatment used in the evidence reviewed, as well as being in line with the suggested use in the BNF and the Summary of Product Characteristics (SPC).
Economic considerationsThe GDG relied on a variety of sources in their consideration of the costs and benefits of alternative topical therapies in the treatment of patients with scalp psoriasis. Limited evidence, both in terms of quantity and quality, was identified in the published literature. One study showed that starting with twice daily betamethasone valerate (potent corticosteroid) followed by concurrent (one applied in the morning and one in the evening) treatment with betamethasone dipropionate (potent corticosteroid) and calcipotriol (vitamin D analogue) and then once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate was the most cost-effective treatment sequence. Due to limitations of the study, the GDG remained uncertain about the robustness of these conclusions.

Original decision modelling was undertaken for the guideline and showed that there were relatively small differences in terms of benefit between different topical sequences for scalp psoriasis, but large differences in terms of cost. Based on the mean costs and benefits of 169 compared sequences, the analysis found that initial treatment with once daily very potent corticosteroids is likely to offer the best value for NHS resource. The GDG was concerned that very potent corticosteroids, although most effective and cost-effective, are quite an aggressive initial strategy and carry greater risk of steroid-related adverse events, which were not captured in the economic model. The second most cost-effective first line treatment in the base case and across a range of sensitivity and scenario analyses was once daily potent corticosteroids. The GDG had noted strong patient preference for once daily applications due to the messiness, inconvenience and cosmetic acceptability of topicals applied to the scalp. Therefore the GDG chose not to recommend once or twice daily very potent steroids as either the first- or second-line treatment. It was considered appropriate as third-line treatment, as the number of patients exposed to the risks would be fewer but the need for efficacy more urgent.

Of the remaining strategies, the most cost-effective strategies were:
  • First-line – once daily potent corticosteroid; second-line – once or twice daily vitamin D or vitamin D analogue; third-line – once or twice daily very potent corticosteroid.
  • First-line – once daily potent corticosteroid; second-line – once daily very potent corticosteroid; third-line – once daily combined product containing calcipotriol monohydrate and betamethasone dipropionate.
Once-daily combined product containing calcipotriol monohydrate and betamethasone dipropionate might only represent a cost-effective second line option after potent corticosteroids alone if patients are expected to use less than 60g per month

The GDG considered it important to think about avoiding the continuous use of corticosteroids (potent or very potent), and on the basis of results from scenario analyses with restricted comparators, found vitamin D or analogue likely to represent the optimal second line choice. However, if a product with steroids was considered necessary and appropriate, they felt once daily TCF product would represent a safer alternative than very potent corticosteroid.

If these topicals fail to bring about control of scalp psoriasis, then the optimal third-line treatment is twice daily very potent corticosteroids. It was considered appropriate as third-line treatment, as the number of patients exposed to the risks would be fewer but the need for efficacy more urgent. The GDG noted strong patient preference for once daily applications due to the messiness, inconvenience and cosmetic acceptability of topicals applied to the scalp. Therefore, if escalation to twice daily very potent corticosteroids was considered unacceptable, then once daily very potent corticosteroid is likely offer the next best value for NHS resource.

The analysis also considered the cost-effectiveness of coal tar polytherapy (Capasal® shampoo) relative to other topicals in the treatment of scalp psoriasis. Coal tar based shampoo was only slightly more effective that placebo/vehicle scalp solution and far less effective than other topicals. In the model, this meant that more patients ended up failing treatment in primary care and being referred for specialist consultations and treatments, thus making the true costs to the NHS of treatment with coal tar shampoos much higher than the acquisition cost alone. The GDG were aware that coal tar based shampoos are regularly prescribed in primary care for treatment of scalp psoriasis and agreed that based on the evidence of clinical and cost-effectiveness that they are not optimal for the treatment of scalp psoriasis. In order to ensure more efficient use of NHS resources, they considered it important to discourage GPs from using this particular treatment modality.

No economic evidence was available to inform the GDG on the relative cost-effectiveness of topicals in the treatment of psoriasis at sites such as the face and flexures. Given the cost-effectiveness of corticosteroids in the treatment of psoriasis of the trunk, limbs and scalp, the GDG concluded that corticosteroids were likely to represent good value for money in the treatment of psoriasis of the face and flexures, if side-effects are manageable. However, they noted the substantial risk of skin atrophy associated with corticosteroid use at these sites, and thus concluded that neither potent nor very potent corticosteroids were safe or appropriate. In the absence of clinical and economic evidence, the GDG relied on their clinical experience with mild and moderate potency corticosteroids. They concluded that their low acquisition cost was very likely to be justified by the benefits gained compared to alternatives. Calcineurin inhibitors are more costly than moderate potency corticosteroids and are not licensed for the treatment of psoriasis. The GDG considered that they may represent good value for NHS resources if continuous treatment is required (and thus the risk of steroid-associated side effects is higher) or if moderate potency corticosteroids fail to bring about the desired level of response.
Quality of evidenceAll studies

The majority of the data on withdrawals (except withdrawals due to lack of efficacy for the placebo comparisons) and skin atrophy across all comparisons showed low event rates that gave very imprecise relative estimates, but in absolute terms demonstrated precise evidence of no clinically relevant difference between the interventions because the numbers involved were so low. Even in cases where there was a statistically significant difference in the interventions, such as withdrawals due to adverse events in the comparison of potent corticosteroids and placebo, in absolute terms there was no clinically significant difference between the interventions.

The study limitations regarding steroid atrophy discussed in relation to trunk and limbs (see 8.5) also apply to high impact and difficult to treat sites.

There was a lack of information regarding the duration of remission/time-to-relapse, which was only reported in 3 studies (Poulin 2010, Klaber 1994 and Kragballe 2009). While there was an overall trend that the relapse rate was higher following use of preparations including potent steroids compared with vitamin D or vitamin D analogues the different definitions of relapse and time-points of assessment made it difficult to assimilate the data.

Scalp psoriasis
  • Vitamin D or vitamin D analogues vs placebo: There was heterogeneity between two studies (Jemec 2008 and Green 1994) included in the comparison of vitamin D or vitamin D analogues vs. placebo for scalp psoriasis for the outcome of investigator’s assessment of achieving clear or nearly clear which was not explained by pre-defined subgroups but may have been due to a higher risk of bias in the Green 1994 study. Nevertheless, both studies suggest that vitamin D or vitamin D analogues are clinically beneficial in terms of achieving clearance or near clearance compared with placebo treatment. It was noted that some patients prefer the solution, as it does not make the hair greasy, which the gel does.
  • Potent corticosteroid vs placebo: One study (Franz 1999) investigating potent corticosteroid vs. placebo on the scalp included two experimental arms with different formulations of active treatment. Although it was not within the review protocol to investigate differences in formulation, the GDG noted that a statistically significant difference was demonstrated between the foam and lotion formulations of betamethasone valerate (foam = 72% response, lotion = 47% response on investigator’s assessment; results for the patient’s assessment were similar).
  • Very potent corticosteroid vs placebo: The study timeframes (Franz 2000, Olsen 1991, Jarratt 2004 and Sofen 2011) for very potent corticosteroid vs. placebo ranged from two to four weeks duration, which may be too short a timeframe to detect skin atrophy. As with potent steroids, foam formulations were more effective than lotion formulations; however the difference was not statistically significant for very potent corticosteroids. One study (Poulin 2010) looked at maintenance of response using very potent steroid vs placebo for up to 6 months but was noted to be of very low quality because once daily clobetasol propionate was permitted for up to 4 weeks if relapse occurred in clobetasol or vehicle group. During the whole study, clobetasol propionate was applied for 79.3 days in the clobetasol propionate group and 59.5 days in the vehicle group.
  • Potent corticosteroids vs vitamin D or vitamin D analogue: There was unexplained heterogeneity between the studies (Jemec 2008, van de Kerkhof 2009 and Klaber 2004) for the efficacy outcomes, but betamethasone dipropionate was clinically beneficial compared to vitamin D or vitamin D analogue treatment.
  • Very potent steroids compared with other active treatments: One study (Reygagne) compared very potent corticosteroid with vitamin D or vitamin D analogue treatment. The skin atrophy treatment effect was unclear because some atrophy was present at baseline. The GDG noted that there were no direct data comparing very potent steroids with other active treatments. However, from the network meta-analysis twice daily very potent corticosteroids were likely to be the most effective treatment. However, once daily potent corticosteroid or combined product containing potent steroid and vitamin D analogue (calcipotriol monohydrate and betamethasone dipropionate) may be more effective than once daily very potent corticosteroid.
  • Combined product containing vitamin D analogue and potent steroid (calcipotriol monohydrate and betamethasone dipropionate) vs. vitamin D or vitamin D analogue alone: There was heterogeneity between the 3 studies (Kragballe 2009, Jemec 2008 and van de Kerkhof 2009) for the outcome of patient’s assessment of scalp clearance comparing a combined product containing calcipotriol monohydrate and betamethasone dipropionate vs. vitamin D or vitamin D analogue alone. This may have been because Kragballe 2009 used a gel formulation of the combined preparation and a solution of vitamin D analogue, so the combination formulation may have been more effective than the vitamin D analogue comparator formulation. All 3 studies suggest that a combined product is clinically beneficial in terms of achieving clearance or near clearance compared with vitamin D or vitamin D analogue treatment alone.
  • Coal tar (shampoo): The GDG commented that the 4–8 week follow-up in the studies (Griffiths 2006A and McKinnon 2000) assessing coal tar to treat scalp psoriasis was too short term to be able to draw any conclusions about the time to maximum effect. It is known from the trunk and limb data that coal tar takes a long time to act. Relapse rate is very low so coal tar probably does have a role in some patients.
    • In relation to different formulations, the GDG agreed that blinding was difficult especially with regard to tar and dithranol.
    • The MacKinnon study was not felt to reflect clinical practice as coal tar shampoos are usually used as an adjunct rather than monotherapy.
Face and flexural (including genital) psoriasis
Overall there are little data for psoriasis at the face and flexural sites, and no data for corticosteroids at these sites. Use of mild to moderate corticosteroids for face and flexural disease is accepted as standard practice and the lack of trial data of sufficient quality to be included in the review is disappointing but may reflect the historical usage. Therefore, based on clinical experience, the GDG agreed to make a recommendation for their use.
Regarding the graphical data for time-to-maximum effect with tacrolimus the findings of the Lebwohl and Liao studies for improvement are conflicting. The Lebwohl study found that the number or people improving after 29 days treatment with tacrolimus was minimal. The Liao study found though that patients with clear/almost clear psoriasis increased by 20% between four and six weeks of treatment. The GDG noted that in the Lebwohl study 0.1% tacrolimus was used compared with 0.3% tacrolimus in the Liao study. Therefore, the differences were thought to be explained by the lower strength formulation taking longer to act.

Scalp, face and flexural (including genital) psoriasis in children
  • The GDG commented on the lack of evidence for the treatment of children with psoriasis at difficult-to-treat sites; although two studies (Jarratt and Reygagne) included ages ≥12, the mean age in both was over 45 years.
  • The GDG agreed that the recommendations for adults could be extrapolated to children and young people for scalp psoriasis provided that healthcare professionals also consulted the relevant SPC and BNF sections. Potent corticosteroids should only be used in those over 1 year of age, in accordance with marketing authorisation.
  • For facial, flexural and genital psoriasis mild and moderate potency corticosteroids were agreed to be appropriate in children as well as adults, however, calcineurin inhibitors were not thought to be appropriate for use in children at the time of publication of this guideline owing to the lack of evidence in children with psoriasis and safety concerns, specifically cancer risk.
Other considerationsThe GDG noted there were no studies that addressed maintenance. As with trunk and limbs, an as-needed approach to use of topicals was appropriate. The point at which treatment should be reinstituted is based on patient need. Return of scale was felt to be significant by patient members of the group.
Scalp psoriasis
  • It is difficult to assess skin atrophy on the scalp.
  • Use of corticosteroid on the scalp can be associated with inadvertent application to the face with consequent risk of skin atrophy, facial acne. Therefore careful application is important.
  • A post-hoc subgroup analysis based on ethnicity (type V and VI skin) for the outcome of investigator’s assessment of clear/nearly in the Tyring 2010 study found no significant difference between the subgroups when comparing the combined product containing calcipotriol monohydrate and betamethasone dipropionate scalp formulation (gel) vs. placebo. However, post-hoc analyses are intrinsically at high risk of bias and the GDG noted that the severity of psoriasis can be underestimated in people with type V and VI skin.
  • Patient preference is an important factor in choosing a formulation to treat scalp psoriasis. The difference in cost of the formulations is small.
  • The majority of the data on withdrawals (except withdrawals due to lack of efficacy for the placebo comparisons) and skin atrophy across all comparisons showed low event rates that gave very imprecise relative estimates, but in absolute terms demonstrated precise evidence of no clinically relevant difference between the interventions because the numbers involved were so low. Even in cases where there was a statistically significant difference in the interventions, such as withdrawals due to adverse events in the comparison of potent corticosteroids and placebo, in absolute terms there was no clinically significant difference between the interventions. The limitations to the studies in relation to steroid atrophy discussed in the trunk and limbs section also apply to high impact and difficult to treat and high impact sites (see 7.4.4 for trunk and limbs).
  • The GDG felt that offering very potent corticosteroids first line would not be appropriate for scalp psoriasis. The GDG were mindful that the treatment is for long term use and relapse rates are higher with very potent steroids. Even use of potent steroid for scalp psoriasis in primary care would be a change in clinical practice. The GDG noted that most of the evidence related to people with moderate-to-severe psoriasis; many people may present for treatment with scaling in the scalp alone and this may be labelled as ‘scalp psoriasis’ and treatment with very potent corticosteroids would not be appropriate. In these individuals coal tar shampoos may be appropriate.
  • From GDG experience, removing the scale on the scalp before applying active treatment improves the efficacy of active treatment.
  • The evidence indicated that coal tar shampoo was of limited benefit. However, the quality of the evidence was poor and limited to one trial only for efficacy outcomes, although coal tar shampoos are very widely used. In addition, the diagnosis of ‘scalp psoriasis’ is sometimes difficult, since it may include other conditions such as seborrhoeic dermatitis, and also only mild scaling in the scalp (in which case use, for example, potent corticosteroids would be inappropriate and coal tar shampoo appropriate). The GDG agreed therefore that a DO NOT use recommendation was only justified in those people with severe disease to ensure that their disease would receive appropriate treatment (ie not coal tar alone).
Face and flexures (including genitals)
  • Calcineurin inhibitors are not prescribed for psoriasis in primary care as they are not licensed to treat psoriasis; however they are licensed and widely used in eczema.
  • The GDG felt that intermittent short-term use of mild or moderately potent corticosteroids could be recommended in primary care but only for short-term use; the use of topical calcineurin inhibitors should be on specialist advice given that these agents are unlicensed.
  • The evidence suggested that for all interventions some level of response should be achieved by 4 weeks in those who are likely to gain benefit; therefore, the GDG agreed that it would be appropriate to review at 4 weeks to assess response to treatment. Additionally, for calcineurin inhibitors, the maximum response appears to be reached by 4 weeks so this was recommended as the treatment duration for this intervention.
  • Non-concordance should be considered if there is no response to treatment in line with ‘Medicines adherence’ (NICE clinical guideline 76)265.
ddd

Only use potent corticosteroids according to UK marketing authorisation, which was limited to those over 1 year of age at the time of publication (October 2012).

eee

In children and young people the specified duration of therapy may not be appropriate. Please refer to the BNF for children for information on appropriate dosing and duration of treatment.

fff

Only use potent corticosteroids according to UK marketing authorisation, which was limited to those over 1 year of age at the time of publication (October 2012).

ggg

In children and young people the specified duration of therapy may not be appropriate. Please refer to the BNF for children for information on appropriate dosing and duration of treatment.

hhh

See recommendation 32 for additional considerations before changing to the next treatment option.

iii

At the time of publication (October 2012), the combined product containing calcipotriol monohydrate and betamethasone dipropionate did not have UK marketing authorisation for this indication in children and young people. The prescriber should follow relevant professional guidance, taking full responsibility for the decision. The patient (or their parent or carer) should provide informed consent, which should be documented. See the General Medical Council’s Good practice in prescribing medicines – guidance for doctors for further information.

jjj

In children, when offering an agent in the vitamin D or vitamin D analogue class choose calcipotriol, because at the time of publication (October 2012) calcitriol and tacalcitol did not have UK marketing authorisation for this group.

kkk

Please refer to the BNF for children for information on appropriate dosing and duration of treatment.

lll

At the time of publication (October 2012), moderate potency corticosteroids did not have UK marketing authorisation for this indication. The prescriber should follow relevant professional guidance, taking full responsibility for the decision. The patient (or their parent or carer) should provide informed consent, which should be documented. See the General Medical Council’s Good practice in prescribing medicines – guidance for doctors for further information.

mmm

In children and young people the specified duration of therapy may not be appropriate. Please refer to the BNF for children for information on appropriate dosing and duration of treatment.

nnn

At the time of publication (October 2012), calcineurin inhibitors did not have UK marketing authorisation for this indication. The prescriber should follow relevant professional guidance, taking full responsibility for the decision. The patient (or their parent or carer) should provide informed consent, which should be documented. See the General Medical Council’s Good practice in prescribing medicines – guidance for doctors for further information.

Footnotes

*

For data on time-to-remission or time-to-maximum effect, absolute time-to-effect data or data from multiple time points in one study were reported as the first preference and graphical data were only included for interventions where such data were not available, or for long-term data not otherwise available. Additionally, data on IAGI, PGA, PAGI or PASI were reported in preference to TSS where available.

Copyright © National Clinical Guideline Centre - October 2012.

Apart from any fair dealing for the purposes of research or private study, criticism or review, as permitted under the Copyright, Designs and Patents Act, 1988, no part of this publication may be reproduced, stored or transmitted in any form or by any means, without the prior written permission of the publisher or, in the case of reprographic reproduction, in accordance with the terms of licences issued by the Copyright Licensing Agency in the UK. Enquiries concerning reproduction outside the terms stated here should be sent to the publisher at the UK address printed on this page.

The use of registered names, trademarks, etc. in this publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant laws and regulations and therefore for general use.

The rights of National Clinical Guideline Centre to be identified as Author of this work have been asserted by them in accordance with the Copyright, Designs and Patents Act, 1988.

Bookshelf ID: NBK327711

Views

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...