This site is intended for healthcare professionals
Plaque psoriasis
Key evidence on psoriasis treatments

Key evidence on psoriasis treatments

Read time: 90 mins
Last updated:6th Aug 2023
Published:6th Aug 2023

Key psoriasis research and clinical data

Psoriasis-related publications have grown substantially since 1960, and recent discoveries have paved the way for future research. Although research topics have expanded and diversified, the goal of research has remained to better understand psoriasis and its treatment to benefit people living with the disease1.

In this section, we provide a concise overview of the key results from the most significant clinical trials for each systemic and biologic treatment currently available.

Fumaric acid esters - key clinical trials in psoriasis

Dimethyl fumarate

The European Medicines Agency's (EMAs) decision to approve a new formulation dimethyl fumarate (DMF) across Europe in 2017 was based on a Phase III, double-blind, placebo-controlled, non-inferiority trial (BRIDGE), which assessed the efficacy and safety of the new formulation DMF, compared with placebo, and an older DMF2

 

The BRIDGE study2

In the BRIDGE trial, patients were randomised to receive a new formulation DMF, an older formulation DMF, or placebo (2:2:1) for 16 weeks, up-titrating to a maximum daily DMF dose of
720 mg, depending upon individual response2.

At Week 16: 

The BRIDGE study at Week 16

LAS410008, a new formulation DMF.

Most treatment‐related adverse events were classed as ‘mild’2.

PDE4 inhibitor key psoriasis clinical trials

Apremilast

Key efficacy and safety findings

ESTEEM 1: Apremilast 30 mg twice daily versus placebo3
  • PASI 75 (Week 16): 33.1% of patients receiving apremilast achieved PASI 75 vs. 5.3% of placebo-treated patients (P<0.0001)
  • PASI 50 (Week 52): 61% of patients re-randomised to apremilast at Week 32 achieved PASI 75 at Week 52 vs. 11.7% re-randomised to placebo
  • Pruritis: ~30% reduction in pruritis visual analogue scale scores
  • PASI 50 in PASI 75 non-responders: ~50% of patients who were PASI 75 non-responders achieved PASI 50 at Weeks 32 and 52 
  • Adverse events (AEs): 69.3% of patients reported at least one adverse event with apremilast vs. 55.7% of placebo-treated patients

 

ESTEEM 2: Apremilast 30 mg twice daily versus placebo (re-randomisation of patients achieving PASI 50)4

ESTEEM 2 study - Apremilast 30 mg twice daily versus placebo

PASI 50 at Week 52: 80% of patients re-randomised to apremilast versus 11.7% re-randomised to placebo achieved PASI 50 at Week 524.

The LIBERATE trial5

The Phase IIIb trial LIBERATE evaluated the efficacy and safety of apremilast vs. placebo in biologic-naïve patients with moderate-to-severe plaque psoriasis, and the safety of switching from etanercept to apremilast. 

It demonstrated that the efficacy of apremilast was sustained over 52 weeks both in patients who continued on apremilast and also in patients who switched from etanercept to apremilast5.

The LIBERATE trial at Week 16

Switching from etanercept to apremilast did not result in any new or clinically significant safety signals.

The UNVEIL trial6

Meanwhile, the Phase IV UNVEIL trial assessed the efficacy and safety of apremilast in systemic- and biologic-naïve patients with moderate plaque psoriasis over 52 weeks. In the initial 16-week randomised, double-blind, placebo-controlled phase of the study, patients receiving apremilast achieved a significantly greater change from baseline in the product of Static Physicians Global Assessment (sPGA) and body surface area (BSA) scores (PGA x BSA) than placebo (–48.1% vs. –10.2%, P<0.0001)6

Patients receiving apremilast also experienced a significantly greater improvement in Dermatology Life Quality Index (DLQI) score than patients given placebo (–4.8 vs. –2.4; P=0.0008)6

UNVEIL extension phase

In the UNVEIL extension phase all patients received apremilast and were followed for up to 52 weeks. Among those patients that completed the study, all efficacy improvements observed at Week 16 were maintained at Week 527.

In a pooled analysis of ESTEEM 1 and 280,81, LIBERATE177, and UNVEIL178, greater achievement of PASI ≤2 was observed in patients with more moderate than more severe skin disease8

TNF-α inhibitor - key clinical trials in psoriasis

Infliximab for psoriasis

Approval of infliximab for psoriasis was based on the two Phase III EXPRESS clinical studies9,10

Key efficacy findings

EXPRESS: infliximab 5 mg/kg vs. placebo9

EXPRESS: IFX 5 mg/kg vs. placebo

EXPRESS II: infliximab 3 mg and 5 mg/kg vs. placebo10

 EXPRESS II: IFX 3 mg and 5mg/kg vs. placebo

PASI scores at Week 50: Better PASI responses were obtained with 5 mg/kg and with continuous maintenance vs. intermittent treatment.

The REALITY health-related quality of life study11

REALITY was a prospective, observational, real-world, open-label study, of infliximab in moderate-to-severe psoriasis that demonstrated substantial improvements in health-related quality of life (HRQoL)11

  • At Week 50, the mean DLQI improved (decreased) from 12.7 at baseline, to 4.7
    • 64% of infliximab patients improved by ≥5 DLQI points
  • At Week 98 mean DLQI was 2.8 
    • 68.8% of infliximab patients improved by ≥5 DLQI points
    • 47.5% of patients achieved a DLQI of 0
The PIECE (Psoriasis Infliximab vs. Etanercept Comparison Evaluation) study12

The PIECE study was a non-pharmaceutical sponsored prospective randomised controlled trial comparing infliximab and etanercept in moderate-to-severe psoriasis.

PIECE demonstrated that although infliximab is associated with a rapid and significantly higher level of short-term efficacy compared to etanercept (at Week 24, PASI 75 was achieved in 72% infliximab vs. 34.8% etanercept, P=0.01), there was no significant difference with longer term maintenance treatment12:

  • At Week 48, PASI 75 was achieved in 66.7% infliximab patients vs. 50% etanercept patients (P=0.65)

Key safety results

Infliximab was generally well tolerated in most patients in the EXPRESS trials. 

Etanercept for psoriasis

Key efficacy findings

Approval of etanercept for psoriasis was based on the results of two Phase III studies described below.

Etanercept low (25 mg weekly), medium (25 mg twice weekly) or high dose (50 mg twice weekly) vs. placebo13

Patients received either:

  • Etanercept low dose (25 mg weekly)
  • Etanercept medium dose (25 mg twice weekly)
  • Etanercept high dose (50 mg high dose)
  • Placebo

Etanercept low (25mg weekly), medium (25 mg twice weekly) or high dose (50 mg twice weekly) vs. placebo - PASI 75 responder rate

At Week 24, PASI 90 was achieved by:

  • 25% of patients receiving etanercept low dose
  • 44% of patients receiving etanercept medium dose
  • 59% of patients receiving etanercept high dose

PASI responses were paralleled by improvements in PGA and QoL. 

Etanercept 25 mg or 50 mg twice weekly vs. placebo14
Etanercept 25 mg or 50 mg twice weekly vs. placebo - PASI 75 responder rate
At Week 24, PASI 75 was achieved by:
  • 54% of patients with dose reduction 50 mg to 25 mg bi-weekly
  • 45% of patients with continuous 25 mg bi-weekly dosing
  • 28% of placebo-treated patients who subsequently received 25 mg bi-weekly dosing

Key safety results

Etanercept was generally well tolerated in the etanercept pivotal clinical trials14

Adalimumab for psoriasis

REVEAL15

In REVEAL, patients received either 40 mg of adalimumab or placebo once every two weeks.

Key efficacy findings

REVEAL - key efficacy findings

Key safety findings

In REVEAL, <2% of patients discontinued the study.

CHAMPION16

In CHAMPION, patients were assigned to receive either adalimumab (80 mg at Week 0, then 40 mg once every two weeks) or methotrexate (7.5–25 mg weekly).

Key efficacy findings 

Key efficacy findings from the CHAMPION study

Key safety findings

In CHAMPION, AEs were similar across treatment groups; discontinuation was greatest with methotrexate.

ESPRIT: long-term observational registry17

ESPRIT is an ongoing, 10-year, observational registry evaluating long-term safety and efficacy of adalimumab in moderate-to-severe psoriasis in routine clinical practice17

Initial (5-year) results demonstrate that efficacy remains stable over five years with no new safety signals17. The study is due to be completed in September 2022.

Certolizumab pegol for psoriasis

Phase III development of certolizumab pegol involved two studies that enrolled approximately 1,000 patients, including patients with and without prior treatment experience with biologic products18,19.

CIMPASI-120

In CIMPASI-1, patients received either placebo or a 400 mg certolizumab pegol (CPZ) loading dose at Weeks 0, 2 and 4, then certolizumab pegol 200 mg or 400 mg every two weeks.

Key efficacy findings

Key efficacy findings from the CIMPASI-1 study

* Secondary endpoint.

Key safety findings

Key safety findings from the CIMPASI-1 study

TEAE, treatment-emergent adverse events.

CIMPASI-220

CIMPASI-2 was a replicate study run in parallel to CIMPASI-1.

Key efficacy findings

Key efficacy findings from the CIMPASI-2 study* Secondary endpoint.

Key safety findings

Key safety findings from the CIMPASI-2 study

TEAE, treatment-emergent adverse events.

CIMPACT21

In CIMPACT, up until Week 16 patients received either:

  • A 400 mg certolizumab pegol loading dose at Weeks 0, 2 and 4, then 200 mg certolizumab pegol every two weeks
  • A 400 mg certolizumab pegol loading dose at Weeks 0, 2 and 4, then 400 mg certolizumab pegol every two weeks
  • Etanercept twice weekly
  • Placebo

Patients who were re-randomised and were PASI 50 non-responders at any visit during the maintenance period were entered into the open-label safety extension and received certolizumab pegol 400 mg every two weeks.

Key efficacy findings

Sustained superiority versus placebo:

Sustained superiority versus placebo - CIMPACT study

Superiority versus etanercept for certolizumab pegol 400 mg every two weeks, and non-inferiority for certolizumab pegol 200 mg every two weeks:

Superiority versus etanercept for certolizumab pegol 400 mg every two weeks, and non-inferiority for certolizumab pegol 200 mg every two weeks

Etanercept PASI 75 responders who were switched to certolizumab pegol or placebo at Week 16:

Etanercept PASI 75 responders who were switched to certolizumab pegol or placebo at Week 16

Etanercept non-responders (<PASI 50 at Week 12) who were switched to certolizumab pegol 400 mg every two weeks:

Etanercept non-responders (<PASI 50 at Week 12) who were switched to certolizumab pegol 400 mg every two weeks

In 3-year results from CIMPASI-1 and CIMPASI-2, at Week 144, PASI 75/90 was achieved by 70.6%/48.7% patients randomised to certolizumab pegol 200 mg, and 72.9%/42.7% randomised to certolizumab pegol 400 mg22.

Key safety findings

Key findings from the CIMPACT study

TEAE, treatment-emergent adverse events.

IL-12 and IL-23 inhibitors – key clinical trials in psoriasis

Ustekinumab (IL-12/23 inhibitor) for psoriasis

Approval of ustekinumab in psoriasis was based on the efficacy and safety data from two Phase III clinical trials, PHOENIX 1 and PHOENIX 2.

Comparable efficacy of ustekinumab (standard dose) has been demonstrated in both adults and psoriasis patients aged 12–17 with no unexpected AEs23.

PHOENIX 124

In this study, ustekinumab was administered at a dose of either 45 mg or 90 mg at Weeks 0 and 4, then every 12 weeks versus placebo. 

Key efficacy findings

Key efficacy findings - PHOENIX 1 study

Key safety findings

During the placebo-controlled phase, AEs and serious adverse events (SAEs) occurred in 54.5%, and 1.2% of ustekinumab-treated patients vs. 48.2% and 0.8% of placebo-treated patients.

PHOENIX 225,26

In this study, ustekinumab was administered at a dose of either 45 mg or 90 mg at Weeks 0 and 4, then every 12 weeks versus placebo. 

Key efficacy findings

Key efficacy findings for the PHOENIX 2 study

Key safety findings

Key safety findings for the PHOENIX 2 study

Ustekinumab vs. other psoriasis treatments (PSOLAR registry data)

Efficacy and safety of ustekinumab and other psoriasis treatments in a real-world setting was assessed using the PSOLAR registry data27,28

Analyses concluded that ustekinumab in people with psoriasis:

  • is more effective than infliximab, etanercept and adalimumab, with no increased risk of malignancy, major adverse cardiovascular events (MACE), serious infection, or mortality27,28
  • presents a lower risk of infections compared to methotrexate treatment (hazard ratio [HR] 0.65; 95% CI, 0.47–0.89)29
  • offers superior drug survival compared with infliximab, adalimumab and etanercept30

The PSOLAR efficacy findings were confirmed by data from the prospective, BioCAPTURE registry comparing one- and five-year efficacy of adalimumab, etanercept and ustekinumab in people with psoriasis in daily clinical practice. BioCAPTURE suggested that after one year, PASI 75 is more often achieved with adalimumab and ustekinumab than etanercept, and that after five years, ustekinumab has the highest efficacy (P=0.019)31.

Guselkumab (IL-23 inhibitor) for psoriasis

VOYAGE-132

In VOYAGE-1, patients received one of three dosing regimens:

  • Guselkumab (GUS) 100 mg at Weeks 0 and 4, and then every 8 weeks thereafter
  • Placebo at Weeks 0, 4 and 12, then guselkumab at weeks 16 and 20, and then every 8 weeks thereafter 
  • Adalimumab (ADA) 80 mg at Week 0, 40 mg adalimumab at Week 1, and then every 2 weeks thereafter

Key efficacy findings

Key efficacy findings for the VOYAGE-1 study

Improvement in DLQI:

  • At Week 16, change in DLQI was
    • –11.2 for guselkumab (P<0.001 vs. placebo; not stated vs. adalimumab)
    • –9.3 for adalimumab
    • –0.6 for placebo
  • At Week 24, change in DLQI was
    • –11.6 for guselkumab (P<0.001 vs. adalimumab)
    • –9.5 for adalimumab
  • At Week 48, change in DLQI was
    • –11.8 for guselkumab (P<0.001 vs. adalimumab)
    • –9.2 for adalimumab
Open-label extension study

VOYAGE-1 continues with an open-label extension study, running from Week 48 to Week 100. At Week 52, all patients began receiving  guselkumab 100 mg every 8 weeks. Patients initially randomised to adalimumab at Week 0 entered a washout period after their final dose of adalimumab at Week 47 and initiated guselkumab 100 mg at Week 52 and every 8 weeks thereafter.

The study is due to be completed in 202233

Overall, PASI 75, PASI 90, and Investigator’s Global Assessment (IGA) response rates at Weeks 52 and 100 remained consistent.

Key efficacy findings from the open label extension study in the VOYAGE-1 trial

Key safety findings

Key safety findings

Drug survival at Week 48:

  • Discontinuation for any reason was 8.5% guselkumab vs. 15.6% adalimumab (P=0.0053)
    • HR 1.88 (95% CI, 1.19–2.98) P=0.007
  • Discontinuation due to lack of efficacy/worsening psoriasis was 0.9% guselkumab vs. 5.1% adalimumab (P=0.0017)
    • HR 5.71 (95% CI, 1.68–19.5) P=0.0054
  • Discontinuation due to AEs was 3.0% guselkumab vs. 2.8% adalimumab (P=0.297)

Key safety findings in the VOYAGE-1 study - drug survival at WEEK 48

In the open-label extension study, rates of AEs, SAEsand infections per 100 patient-years were consistent with those recorded for VOYAGE-1 up to Week 48.

VOYAGE-234

The VOYAGE-2 study addressed whether benefits observed in patients who responded to guselkumab would be maintained following withdrawal34

Key efficacy findings

Clinical outcomes were significantly better in patients who were maintained with treatment compared with those who received placebo.

Key efficacy findings for the VOYAGE-2 study

VOYAGE-2 efficacy and safety at 4 years

Results of continuous guselkumab treatment through 4 years from VOYAGE 2 revealed efficacy was maintained through 4 years of continuous guselkumab treatment for moderate-to-severe psoriasis, and comparable with patients who crossed over from adalimumab to guselkumab35. Guselkumab showed a favourable safety profile through 4 years, with no new safety signals observed35.

PASI Scores at Week 204

Almirall_PA_MarUpdate.KI5.png

Patient‐Reported Outcome Scores at Week 204

Almirall_PA_MarUpdate.KI6.png

GUS, patients receiving GUS 100 mg every 8 weeks. A>GUS, ADA non-responders switched to GUS 100 mg, receiving GUS at or after Week 28.

VOYAGE-1 and 2 - 5-year efficacy and quality of life

In 5-year results, the proportions of patients in the guselkumab group who achieved clinical responses at Week 252 in VOYAGE 1 and VOYAGE 2, respectively, were 84.1% and 82.0% (≥90% improvement in PASI; 82.4% and 85.0% (IGA 0 or 1); 52.7% and 53.0% (100% improvement in PASI), and 54.7% and 55.5% (IGA 0)36.

Health-related QoL (HRQoL) endpoints were 72.7% and 71.1% of patients (DLQI 0 or 1: no effect on patient’s life); 42.4% and 42.0% (Psoriasis Symptoms and Signs Diary [PSSD] symptom score=0), and 33.0% and 31.0% (PSSD sign score=0). As measured in VOYAGE 2 only, approximately 45% of patients achieved ≥5-point reduction in Short Form-36 physical and mental component scores, and 80% reported no anxiety or depression (Hospital Anxiety and Depression Scale scores <8). Similar findings were reported for adalimumab crossovers. These effects were maintained from Week 52 in VOYAGE 1 and Week 100 in VOYAGE 236.

Key safety findings at Week 204

Almirall_PA_MarUpdate.KI7.png

In 5-year safety results from VOYAGE-1 and 2, AEs rates were low, and generally remained stable over time with continuous guselkumab exposure through 5 years37.

ECLIPSE34

ECLIPSE is the first head-to-head study to compare efficacy between guselkumab and secukinumab33

Patients received either:

  • Guselkumab 100 mg by subcutaneous injection at Weeks 0 and 4, followed by a maintenance dose every 8 weeks, or
  • Secukinumab 300 mg (2 subcutaneous injections of 150 mg) at Weeks 0, 1, 2, 3 and 4 followed by monthly maintenance dosing

ECLIPSE study

Key safety findings

Safety profiles were consistent with the known safety profiles seen in the respective registration trials and the current prescribing information.

POLARIS38

A completed Phase IIIb national, multicentre, randomised, open-label, assessor-blinded, active comparator trial (POLARIS; NCT02951533) compared the efficacy of guselkumab to fumaric acid esters (FAE) in people with moderate-to-severe psoriasis who are candidates for, and naïve to, systemic treatment in Germany38.

In this study, 82% of patients receiving guselkumab achieved PASI 90 versus 14% receiving FAE at Week 24 (P<0.001)38.

While 28% of patients receiving FAE discontinued due to AEs, no patients receiving guselkumab had to discontinue for this reason38.

Tildrakizumab (IL-23 inhibitor) for psoriasis

In Phase III studies outlined below, tildrakizumab was associated with significantly higher proportions of people with moderate-to-severe psoriasis achieving PASI 75 and clear or minimal PGAs than placebo.

ReSURFACE long-term data – expert opinion

Watch the short video clips, featuring Professor Diamant Thaçi (Director, Institute and Comprehensive Center for Inflammation Medicine, University of Lübeck, Germany), to learn more about tildrakizumab and why long-term safety and efficacy data for biologics is so important.

Why is long-term efficacy and safety biologics data important for treating psoriasis?
In the ReSURFACE psoriasis study, were new safety signals evident only after long-term use of tildrakizumab?
In the ReSURFACE psoriasis study, how do the IL-23p19 inhibitors compare in the real-world population?
reSURFACE 139

In reSURFACE 1, patients were randomised to one of the following at Weeks 0 and 4, and then every 12 weeks thereafter they received tildrakizumab 100 mg or 200 mg, or placebo.

Placebo-treated patients were re-randomised to tildrakizumab at either 100 mg or 200 mg at Weeks 12 and 16. Dosing continued to Week 64.

Key efficacy findings

reSURFACE 1 key efficacy findings

ReSURFACE 2 (tildrakizumab versus etanercept)

ReSURFACE 2 also showed that tildrakizumab 200 mg was associated with significantly higher proportions of patients achieving PASI 75 and PGA responses at Week 12 than etanercept, an effective anti-TNF-α treatment for psoriasis39

In patients who were non-responders or partial responders to etanercept in the reSURFACE 2 study, who joined a 2-year extension phase, tildrakizumab 200 mg resulted in PASI 75 responses in almost 9 out of 10 patients after 3 doses and these responses were maintained at Week 14840.

ReSURFACE 2 responder rates and clear or minimal PGAReSURFACE extension (long-term response of tildrakizumab)

All patients enrolled on an extension study achieved at least PASI 50.

In the long-term extension to reSURFACE 1 and reSURFACE 2, PASI 75 and PASI 100 were maintained at Week 148 in etanercept non- or partial responders who were switched to tildrakizumab 200 mg40. Tildrakizumab demonstrated maintenance efficacy for at least 3 years:

Tildrakizumab demonstrated maintenance efficacy for at least 3 years - reSURFACE extension study

ReSURFACE 1 and 2 five-year efficacy and safety data41

Pooled from the reSURFACE 1 and reSURFACE 2 Phase III studies, tildrakizumab is the first anti-interleukin (IL)-23p19 treatment for which five-year efficacy and safety data are reported for patients with moderate-to-severe psoriasis who respond at week 2841.

Efficacy analyses included 329 and 227 Week 28 responders to tildrakizumab 100 mg and tildrakizumab 200 mg, respectively, and 121 etanercept partial responders/non-responders switched to tildrakizumab 200 at Week 28. Safety was assessed from AEs in all treated patients41.

Key efficacy findings

Almirall_PA_MarUpdate.KI1.png

Key safety findings

Almirall_PA_MarUpdate.KI2.png

Notable adverse events

Almirall_PA_MarUpdate.KI3.png

Risankizumab (IL-23 inhibitor) for psoriasis

UltIMMa-142

Patients were randomised to either:

  • Risankizumab (RIS) 150 mg at baseline, and then at Weeks 4, 16, 28, and 40
  • Ustekinumab (UST) 45 mg or 90 mg (based on screening weight) at baseline, then at Weeks 4, 16, 28, and 40
  • Placebo

At Week 28, patients who achieved sPGA 0/1 were re-randomised to either:

  • Risankizumab (maintenance)
  • Placebo (withdrawal)

Treatment continued until Week 104; however, from Week 32, patients who relapsed (sPGA ≥3) were re-treated with risankizumab.

Key efficacy findings

ultIMMa-1 responder rates

Key safety findings

From baseline to Week 16, at least one adverse event occurred in:

  • 49.7% of risankizumab-treated patients
  • 50.0% of ustekinumab-treated patients
  • 51.0% of placebo-treated patients

Through to Week 16, SAEs occurred in:

  • 2.3% of risankizumab-treated patients
  • 8.0% of ustekinumab-treated patients
  • 2.9% of placebo-treated patients
UltIMMa-242

A replicate study of ultIMMa-1.

ultMMa-2 - responder rates

IMMhance43

Patients were randomised to either:

  • Risankizumab 150 mg at baseline, at Week 4 and then every 12 weeks thereafter
  • Placebo

At Week 28, patients who achieved sPGA 0/1 were re-randomised to either:

  • Risankizumab (maintenance)
  • Placebo (withdrawal)

Treatment continued until Week 104; however, from Week 32, patients who relapsed (sPGA ≥3) were retreated with risankizumab.

IMMhance - responder rates

At Week 52, 87% of risankizumab-treated patients (maintenance group) achieved sPGA 0 or 1, versus 61% of the maintenance group.

Key safety findings

From baseline to Week 16, at least one adverse event occurred in:

  • 45.5% of risankizumab-treated patients
  • 48.0% of placebo-treated patients

Through to Week 16, SAEs occurred in:

  • 2% of risankizumab-treated patients
  • 8% of placebo-treated patients
IMMvent44

Patients were randomised to either:

  • Risankizumab 150 mg at baseline, at Week 4, and then every 12 weeks thereafter
  • Adalimumab (ADA) 80 mg at baseline, followed by 40 mg every other week starting at Week 1

Dose modification at Week 16:

  • Adalimumab-treated patients with a <50 PASI response were switched to risankizumab
  • Adalimumab-treated PASI 90 responders continued with adalimumab
  • Adalimumab-treated PASI 50 responders with a <90 PASI response were re-randomised to risankizumab or adalimumab 

 

IMMvent - responder rates

Key safety findings

From baseline to Week 16, at least one adverse event occurred in:

  • 55.8% of risankizumab-treated patients
  • 56.9% of adalimumab-treated patients

Through to Week 16, SAEs occurred in:

  • 3.3% of risankizumab-treated patients
  • 3.0% of adalimumab-treated patients

IL-17 inhibitors - key clinical trials in psoriasis

Ixekizumab for psoriasis

UNCOVER-145

Patients received either:

  • Ixekizumab (IXE) 80 mg every two weeks 
  • Ixekizumab 80 mg every four weeks 
  • Placebo

UNCOVER-1 - responder rates

UNCOVER-2 and UNCOVER-346

Patients received either:

  • Ixekizumab 80 mg every two weeks 
  • Ixekizumab 80 mg every four weeks
  • Etanercept 
  • Placebo

 In the UNCOVER trials, ixekizumab treatment also resulted in46:

  • High levels of response in etanercept non-responders
  • High efficacy in biologic naïve and experienced patients
  • Greater improvements in nail psoriasis versus etanercept and placebo
  • Sustained improvement in scalp psoriasis versus etanercept and placebo
  • Significant and clinically meaningful improvements in itch (P<0.001)

Key safety findings

SAEs were observed in:

  • 1.9% of patients receiving ixekizumab 2 weekly
  • 1.9% of patients receiving ixekizumab 4 weekly
  • 1.9% of patients receiving etanercept
  • 1.9% of patients receiving placebo (P<0.0001)

The safety profile at Week 60 was similar to that at Week 12.

IXORA-S47

Patients received either:

  • Ixekizumab 160 mg given as a loading dose, followed by 80 mg at Week 2, 4, 6, 8, 10, and 12, and 80 mg ixekizumab every four weeks thereafter
  • 40 mg or 90 mg ustekinumab at Weeks 0 and 4, then every 12 weeks thereafter

IXORA-S - responder rates

Head-to-head studies

IXORA-R (ixekizumab versus guselkumab)48,49

The IXORA-R head-to-head trial, designed to evaluate superiority between ixekizumab and guselkumab was the first head-to-head trial between an anti-IL-17 and anti-IL-23 therapy using the PSAI 100 score as a primary endpoint.

The results showed that:

  • The primary end point of PASI 100 at Week 12 was achieved in 41% of patients in the ixekizumab group compared to 25% with guselkumab (P<0.001)
  • Serious adverse event frequency was 3% for each group with no new safety signals identified
  • At Week 24, ixekizumab was noninferior to guselkumab (50% vs. 52%, respectively) with no significant difference in PASI 100 (P=0.41)
Ixekizumab versus fumaric acid esters and methotrexate

The efficacy of ixekizumab was compared to fumaric acid esters (FAEs) and methotrexate in a head-to-head Phase III trial of 162 patients with moderate-to-severe psoriasis who were naïve to systemic treatment50. Patients were randomised 1:1:1 to ixekizumab, FAE or methotrexate for 24 weeks of treatment50. The primary outcome was the proportion of patients who achieved PASI75, while key secondary outcomes included 24-week PASI 90 and 100, static Physician’s Global Assessment (sPGA) score (0 or 1), and Dermatology Life Quality Index (DLQI) score (0 or 1)50.

At week 24, the results showed50:

  • Significantly more patients treated with ixekizumab achieved PASI75, PASI 90 and PASI 100 than those who received FAEs or methotrexate (91% vs 22% vs 70%; 80% vs 9% vs 39%; 41% vs 4% vs 13%, respectively)
  • Results for sPGA (0,1) in patients with baseline sPGA ≥3 and DLQI (0,1) were superior for ixekizumab versus methotrexate or FAEs
  • The safety profile of all treatments were consistent with previous studies

Bimekizumab for psoriasis

Bimekizumab received EMA approval for moderate-to-severe psoriasis in 202151. Overall, the safety and efficacy of bimekizumab has been evaluated in patients with moderate-to-severe plaque psoriasis across four Phase III multicentre, randomised, placebo and/or active comparator-controlled clinical trials51–56.

  • In BE READY (n = 435), more participants treated with bimekizumab achieved PASI90 (91%) and Investigator’s Global Assessment (IGA) success (response of 0 or 1 in 93%), than those who received placebo (1% for both; P<0.0001 for both) after 16 weeks55
  • In BE VIVID (n = 567), more participants treated with treated with bimekizumab (85%) achieved PASI90 than those who received either ustekinumab or placebo (50% and 15%, respectively; P<0.0001 for both); and IGA success (84%) versus ustekinumab and placebo (53% and 5%, respectively; P<0.0001 for both) after 16 weeks56
  • In BE SURE (n = 478), more participants treated with bimekizumab (86.2%, pooled analysis) achieved PASI90, than those who received adalimumab (47.2%, P<0.001); and IGA score of 0/1 (85.3%, pooled analysis) versus adalimumab (57.2%, P<0.001)54
  • In BE RADIANT (n = 743), more participants treated with bimekizumab (61.7%) achieved PASI100, than those who received secukinumab (48.9%, P<0.001) after 16 weeks53

Efficacy of bimekizumab was maintained for up to 56 weeks of treatment in BE READY, BE RADIANT and BE VIVID52,53,55,56.

In the BE READY trial, AEs were reported more frequently in participants who received bimekizumab than those who received placebo up to week 16 (61% versus 41%)55. The most common AEs with bimekizumab were nasopharyngitis (9%), oral candidiasis (9%) and upper respiratory tract infections (3%)55. In a meta-analysis of six randomised bimekizumab trials, pooled analysis revealed no obvious differences in treatment-emergent AEs (TEAEs) and serious TEAEs between the bimekizumab and placebo groups (RR 1.17; 95%CI 0.93–1.47; P>0.05; and RR 0.67; 95%CI 0.28–1.61; P>0.05, respectively)57.

In the head-to-head trials, similar rates of AEs were reported for bimekizumab and secukinumab up to 48 weeks (86.1% versus 81.4%)53), adalimumab up to 24 weeks (70.9-72% versus 69.8%)54), and ustekinumab up to 16 weeks (56% vs 51%)56.

Brodalumab for psoriasis 

Phase III development for brodalumab consists of one placebo-controlled trial (AMAGINE-1) and two head-to-head studies comparing brodalumab (BROD) to ustekinumab (AMAGINE-2 and 3)58,59. Based on the results of the AMAGINE studies, brodalumab was submitted to regulatory authorities in Europe and the USA in late 2015/early 2016.

AMAGINE-158                                                           

Patients received either:

  • Brodalumab 140 mg 
  • Brodalumab 210 mg 
  • Placebo

AMGINE-1 - responder rates

AMAGINE-2 and AMAGINE-359

During the induction phase, patients received either:

  • Brodalumab 140 mg 
  • Brodalumab 210 mg 
  • Ustekinumab 45 mg or 90 mg (weight-based dosing)

Following the induction phase, patients receiving brodalumab at Week 12 were re-randomised to receive either:

  • Brodalumab 210 mg every two weeks
  • Brodalumab 140 mg every two weeks
  • Brodalumab 140 mg every four weeks
  • Brodalumab 140 mg every eight weeks

Patients who originally received placebo were switched to brodalumab every two weeks. 

At Week 52, those patients still receiving ustekinumab were switched to brodalumab 210 mg every two weeks for the extension phase.

AMAGINE-2 and AMAGINE-3 - responder rates

PASI 100 was achieved in 50% of patients at 14 weeks for brodalumab and at 44 weeks for ustekinumab.

AMAGINE-2 long-term extension study60

Efficacy data was reported for patients who received brodalumab 210 mg every two weeks through to Week 120 of the long-term extension phase. 

AMAGINE-2 long-term extension study - responder rates

Secukinumab (IL-17 inhibitor) for psoriasis

ERASURE61

Patients received either 150 mg or 300 mg secukinumab (SEC) (once weekly for 5 weeks, then monthly) or placebo.

ERASURE - responder rates

In addition:

  • The proportion of patients achieving PASI 75 and IGA 0/1 increased to Week 16 and was maintained 
  • Secukinumab 150 mg and 300 mg was superior to placebo with respect to the PASI 90 and PASI 100 responses at Week 12 (P<0.001)
FIXTURE61

Patients received either:

  • Secukinumab 150 mg or 300 mg (once weekly for 5 weeks, then monthly) 
  • Etanercept (ETN) 50 mg (twice weekly for 3 months, then once weekly)
  • Placebo

FIXTURE - responder rates

In addition, rates of PASI 90 and PASI 100 were higher among patients receiving secukinumab than those receiving etanercept at Week 12 to Week 52 (P<0.001).

Additional analyses found that efficacy obtained at week 16 was sustained for 2 years.

FEATURE62

Patients received either:

  • Secukinumab 150 mg or 300 mg (once weekly for 5 weeks, then monthly) 
  • Etanercept 50 mg (twice weekly for 3 months, then once weekly)
  • Placebo

FEATURE - responder rates

100% of subjects successfully self-administered treatment at Week 1.

JUNCTURE63

Patients received either:

  • Secukinumab 150 mg or 300 mg 
  • Placebo

JUNCTURE - responder rates

IL-36 inhibitor - key clinical trials in psoriasis

Spesolimab for psoriasis

The key trial that supported the EMA’s conditional approval of spesolimab for generalised pustular psoriasis was a randomised, double-blind, placebo-controlled Phase II study (Effisayil 1), conducted in 53 adult patients with flares of generalised pustular psoriasis64.

In this study, 53 patients were randomised to receive a single intravenous dose of spesolimab 900 g or placebo. Both groups could receive an open-label dose of spesolimab on day 8 or as a rescue medication after day 8, or both, and were followed to week 12.

At the end of week 1:

  • A total of 54% of patients in the spesolimab group achieved the primary endpoint, which was a Generalised Pustular Psoriasis Physician Global Assessment (GPPGA) pustulation subscore of 0 (no visible pustules), compared with 6% in the placebo group (P<0.001)
  • A total of 43% in the spesolimab achieved the secondary end point of 0 or 1 (clear or almost clear skin) as their GPPGA total score, compared with 11% in the placebo group (P=0.02)

The treatment effect was observed irrespective of the mutation status of IL-36RN, the gene that encodes IL-36 receptor inhibitor64,65.

Key safety findings

Within the first week after treatment, AEs were reported in 66% of patients who received spesolimab, versus 56% in the placebo group; while 29% of participants in the spesolimab group had AEs that were considered by the investigator as drug-related, compared to 28% in the placebo group 65. By the end of the first week, infections were reported in 17% of participants in the spesolimab group versus 6% in placebo, and in this reached 47% of participants at week 1265.

At the end of week 1, serious AEs (SAEs) had been reported in 6 participants treated with spesolimab and none in the placebo group 65. Of these SAEs, drug reactions with eosinophilia and systemic symptoms (DRESS) were reported in 2 patients treated with spesolimab, one of which occurred at the same time as a drug-induced hepatic injury65.

Overall, results of this study indicate spesolimab is associated with infections and systemic drug reactions 65. However, data from larger and longer term trials of spesolimab are needed in patients with pustular psoriasis 65.

TYK2 inhibitor – key clinical trials in psoriasis

Deucravacitinib

The 2023 EMA approval of deucravacitinib for the treatment of moderate-to-severe plaque psoriasis66,67 was based on the efficacy and safety results of two multicentre, randomised, double-blind trials: POETYK PSO-1 and POETYK PSO-2 67–70.

In both trials, patients with moderate-to-severe plaque psoriasis were randomised to deucravacitinib 6 mg once daily, apremilast 30 mg twice daily, or placebo67–69.

  • Participants assigned to placebo were switched to deucravacitinib at week 16, which was continued up to week 52 67–69
  • Participants assigned to apremilast who did not achieve a PASI 50 (POETYK PSO-1) or PASI 75 (POETYK PSO-2) response at week 24 were switched to deucravacitinib, continued until week 52 67–69.
  • For the deucravacitinib arms:
    • In POETYK PSO-1, patients who were randomised to deucravacitinib continued treatment up to week 5267,68
    • In POETYK PSO-2, patients who were randomised to deucravacitinib and achieved PASI 75 at week 24 were re-randomised 1:1 to continue deucravacitinib (maintenance) or were switched to placebo (withdrawal) 67,69

In both studies, the co-primary endpoints were the proportions of patients who achieved the following at week 16 67–69:

  • ≥75% improvement in PASI scores (PASI 75) from baseline, and
  • a static Physician’s Global Assessment (PGA) score of clear or almost clear (0 or 1) versus placebo

Key results for the co-primary endpoints are summarised in Table 1.

Table 1. Results for co-primary endpoints at week 16 from the POETYK PSO-1 and POETYK PSO-2 trials 67–69. PASI75, ≥75% reduction from baseline in Psoriasis Area and Severity Index; sPGA, static Physician’s Global Assessment.
*P≤ 0.0001 for comparison between deucravacitinib and placebo or deucravacitinib and apremilast. Co-primary endpoint comparing deucravacitinib with placebo. p <0.001 for comparison between deucravacitinib and apremilast.

  PASI 75
n (%)
sPGA score of 0 or 1
n (%)
POETYK PSO-1
Deucravacitinib (n = 332) 194 (58.4) 178 (53.6)
Apremilast (n = 168) 59 (35.1)* 54 (32.1)*
Placebo (n = 166) 21 (12.7)*† 12 (7.2)*†
POETYK PSO-2
Deucravacitinib (n = 511) 271 (53.0) 253 (49.5)
Apremilast (n = 254) 101 (39.8) 86 (33.9)*                    
Placebo (n = 255) 24 (9.4)*† 22 (8.6)*†

In both POETYK PSO-1 and PSO-2 trials, PASI 75 response was maintained for up to 52 weeks in the majority of patients (≥80%) who continued deucravacitinib treatment67–69.

Key safety findings

Deucravacitinib was considered well tolerated68, and adverse event (AE) rates were considered similar to those with placebo and apremilast across the POETYK PSO-1 and POETYK PSO-2 trials68,69. The most frequent AEs in the deucravacitinib group were nasopharyngitis and upper respiratory tract infection up to week 52 68,69.

However, across both trials, there was a higher incidence of serious infections and herpes zoster infections up to week 52 in the group that received deucravacitinib, compared with those who received apremilast or placebo68,69. All cases of herpes zoster were mild to moderate in severity, localised and did not lead to discontinuation68,69.

Comparative data on treatments for psoriasis

There are a limited number of head-to-head trials that have directly compared efficacy and safety profiles between psoriasis treatments71,72. These include Phase III comparisons of the efficacy ± safety profiles between71,72:

  • TNF-α inhibitors versus IL-17, IL-12/23 or IL-23 inhibitors
  • TNF-α inhibitors versus the PDE4 inhibitor, apremilast
  • IL-12/23 inhibitor versus IL-17 inhibitors
  • IL-23 inhibitors versus IL-17 inhibitors
  • Types of TNF-α inhibitors

In addition, several systematic reviews and network meta-analyses have also indirectly compared the relative efficacy of treatments for moderate-to-severe psoriasis72–79. Results of indirect comparisons are considered to fill an important evidence gap, as it is not feasible to undertake head-to-head comparisons of all treatments for psoriasis80.

For example, Armstrong and collagues (2021) incorporated results of the head-to-head IXORA-R, ECLIPSE, CLARITY, IMMerge clinical trials into a network meta-analysis, which assessed short-term and long-term PASI response rates across trials of FDA- and EMA-approved treatments in moderate-to-severe plaque psoriasis72. This analysis indirectly compared results across Phase II–IV randomised controlled trials, using a Bayesian network meta-analysis approach to estimate PASI 75/90/100 response rates from treatment in the short-term (i.e. 10–16 weeks from baseline) or long-term (i.e. 48–52 weeks from baseline)72. Compared to other approved treatments for moderate-to-severe psoriasis, results of this meta-analysis indicated72:

  • PASI response rates were highest for ixekizumab, risankizumab and brodalimumab in the short-term (n=71 trials)
  • PASI response rates were highest for risankzumab in the long-term (n=11 trials)

However, it is important to note the results of this indirect comparison (and others) are limited by differences in study design, conduct and patient populations across trials included in the analysis72 .

Also of note, rankings for most biologic treatments for psoriasis have been reported to vary across indirect comparisons80. This may be due to differences in methodologies for comparisons, variations in treatments that were included in each analysis, as well as treatment dosing, during, outcome definitions and effect measures between indirect comparisons80

When considering results and the clinical relevance of indirect comparisons, treatment rankings should not be considered in isolation, and rather should be considered alongside actual differences in outcomes80

Further head-to-head trials are required to more accurately rank psoriasis treatment options according to their risk/benefit ratio71, and more long-term analyses are needed to better understand the comparative long-term efficacy of treatment options for psoriasis and inform clinical decision-making 80.

Read about real-world evidence for biologics in psoriasis

Explore effective treatment selection in psoriasis

Real-world evidence for biologics in psoriasis

Real-world evidence on psoriasis treatments complement data from clinical trials, providing evidence on safety and effectiveness of approved psoriasis treatments in broader populations outside those permitted by the inclusion/exclusion criteria of clinical trials81. Derived from observational, cohort and registry data from everyday clinical practice, real-world evidence for psoriasis includes data from patients with multiple comorbidities, multiple prior treatment failures, and the elderly81–84.

Expert opinion on real-world evidence for psoriasis

Join Professor of Dermatology, Ulrich Mrowietz, in this discussion on the real-world evidence for the efficacy and safety of biologic treatments for psoriasis.

  • Compare phase III clinical trial and real-world evidence data for psoriasis
  • Evaluate if biologic treatments meet patient and practitioner expectations

Does real-world data demonstrate comparative efficacy and safety to phase III clinical trial data when evaluating biologic treatments?

Does real-world data inform us about biologic drug survival and relapse rates in people with psoriasis?

Is real-world evidence for biologic treatment efficacy sufficient to aid practitioners and patients in shared decision making?

Key findings from real-world evidence on biologics for psoriasis

Cohort and registry data have confirmed the real-world effectiveness and safety profile of various biologics for psoriasis, and have provided further information on the association between treatment history and patient characteristics with treatment response, and potential safety risks.

Numerous real-world studies have confirmed the effectiveness and safety profile of various IL-1785–87, IL-2388, IL-12/2389–92 and TNF-α inhibitors 83,93 in patients with psoriasis in clinical practice. These real-world populations include patients with comorbidities, multiple prior treatment failures, and the elderly.

Key findings include:

  • Response rates in the real-world can be lower when compared with data from clinical trials83,94
  • Higher response rates have been reported in patients who are biologic-naive, compared with those who are biologic-experienced84,87,94

In addition, registry data have also been used as a basis to analyse biologic effectiveness according to prior biologic exposure95, and the effectiveness of within-class switches between biologics (e.g. for IL-17 inhibitors96) in patients with psoriasis.

Read on to learn more about key safety findings from real-world evidence, as well as evidence for use of biologics in specific populations, and comparative data between different types of biologics.

Key real-world safety findings for biologics in psoriasis

Risk of infections

Clinical trials that indicated an increased incidence of candidiasis in patients treated with IL-17 inhibitors, and real-world safety data from WHO, EMA, registry and cohort populations also support an association between treatment with IL-17 inhibitors and an increased risk of oropharyngeal, esophageal and cutaneous candidiasis97.

In addition, results of a retrospective cohort study of patents with psoriasis or psoriatic arthritis (n = 9305) in the US indicated 98:

  • the risk of serious infections in biologic-naive patients treated with IL-12/23 inhibitors was lower than those treated with TNF-α or IL-17 inhibitors
  • no difference in infection across TNF-α, IL-17 or IL-12/23 inhibitors in biologic-experienced patients

Risk of IBD

Phase III clinical trials have indicated an association between IL-17 inhibitors treatment and exacerbation or new-onset inflammatory bowel disease (IBD) in patients with psoriasis99. However, results of a nationwide, population-based study using the French National Health Data System has indicated no increased risk of developing IBD in patients with psoriasis and psoriatic arthritis/ankylosing spondylitis who initated IL-17 inhibitor treatment, compared with etanercept99. A small study of the French national registry (MISSIL) also reported that new-onset IBD events were uncommon with IL-17 inhibitor treatment in patients with psoriasis or spondyloarthritis100.

Risk of tuberculosis reactivation

Guidelines recommend screening for tuberculosis (TB) prior to starting treatment with a biologic101–103.

In patients with psoriasis and latent TB, use of TNF-α inhibitors has been associated with an increased risk of TB reactivation across both clinical trials and real-world studies103,104. However, the incidence of conversion to positive TB in real-world patients with psoriasis treated with IL-17, IL-23 and IL-12/23 inhibitors appears to be low105–108, although there is limited long-term data available106.

In a pooled cohort study of over 12,000 patients with psoriasis, psoriatic arthritis or ankylosing spondylitis, spontaneously reported new latent TB infection during secukinumab treatment was rare106. No cases of active TB or latent TB activation were reported106.

In an Italian retrospective chart review of 59 patients with moderate-to-severe psoriasis who were positive for TB and treated with secukinumab, no cases of TB reactivation were reported after up to 84 weeks of treatment105. This included patients who did not receive prior prophylaxis for TB, and no unexpected safety signals were reported105.

Biologics in specific patient populations

Older patients with psoriasis

Considering older patients are often excluded from clinical trials, a study of the Netherlands BioCAPTURE registry compared drug survival (i.e. the probability of remaining on therapy), safety and effectiveness of biologics in older (≥ 65 years) versus younger patients (<65 years) with psoriasis82. In the drug survival analysis, treatment discontinuation due to ineffectiveness, AEs, ineffectiveness and AEs combined, other reasons, and death were considered events82.

In this cohort, ~65% were biologic-naive (~65%), while around ~20% had used 1 previous biologic and ~8% had used 2 previous biologics. Overall,  ~66% were on TNF-α inhibitors, 22.8% on the IL-12/23 inhibitor ustekinumab, and the remaining ~11% were on IL-17 inhibitors82.

After correcting for confounders, similar trends were observed in the drug survival outcomes between the older and younger patient groups82.

Results of a BioCAPTURE registry study indicated biologics were well tolerated and effective in older patients; although they reported discontinuing biologics more often because of ineffectiveness compared with younger patients, no clear differences in PASI were observed between the two groups82In addition, treatment discontinuation due to AEs were no more frequent in older patients compared to younger patients82.

Asian versus non-Asian patients with psoraisis

An analysis of the US and Canadian CorEvitas Psoriasis Registry compared the 6-month effectiveness of biologic therapy between Asian (n = 293) and non-Asian patients with plaque psoriasis (n = 2,314)109, using PASI response, disease activity measures and patient-reported outcomes. After adjustment for confounders, results were similar between groups, except for Dermatology Life Quality Index (DLQI) – there was a 33% lower odds of achieving a DLQI score of 0/1 in Asian versus non-Asian patients109.

Although results of a CorEvitas Psoriasis Registry study indicated that responses to biologic treatment for psoriasis were similar between Asian and non-Asian patients, DLQI analysis indicated Asian patients may experience lesser responses in skin-related quality of life after biologic treatment, when compared to non-Asian patients with psoriasis109

Real-world comparative data on biologics in psoriasis

Comparative effectiveness of TNF-α inhibitors, IL-17 and IL-23 inhibitors

The first-year effectiveness of etanercept, adalimumab, ustekinumab, secukinumab, ixekizumab and guselkumab was compared across 700 patients with psoriasis in a prospective, multicentre cohort study using data from the BioCAPTURE registry83. In this cohort, >30% had psoriatic arthritis, and ~62% were biologic-naive.

Based on PASI 90 and PASI75 data (correcting for baseline PASI, biologic naivety and weight), results indicated83:

  • Patients treated with adalimumab, ustekinumab, secukinumab, ixekizumab or guselkumab had significantly higher overall odds of reaching PASI90 than those treated with etanercept
  • Patients treated with ixekizumab or guselkumab also had higher probabilities of reaching PASI90 than those treated with adalimumab, ustekinumab or secukinumab

The authors concluded that ixekizumab and guselkumab showed high effectiveness in daily practice, whereas etanercept appeared to be least effective83.

In this cohort study based on BioCAPTURE Registry data, a 90% reduction in baseline PASI was reached less often in this real-world population with TNF-α, IL-17, IL-12/23 and IL-23 inhibitors than that expected based on results from randomised controlled trials83

Comparative drug survival

In real-world studies, drug survival (i.e. the probability of remaining on therapy) is often used as a proxy measure for effectivess84. It reflects the time from initiation to discontinuation of a treatment, and helps to predict the likelihood of a patient remaining on a specific treatment110.

Numerous real-world drug survival studies have demonstrated biologic-naive patients are more likely to have an adequate response to treatment, and have higher drug survival when compared with biologic-experienced patients84.

In patients with psoriasis, drug survival rates at one year and after four years were 69% and 47% respectively for adalimumab, and 82% and 56% for ustekinumab84. Drug survival for IL-17 inhibitors ixekizumab, guselkumab and secukinumab has been reported as 75%, 80% and 76%, respectively, after one year, and can vary with biologic experience84.

In a retrospective, multi-country study of 4,187 patients with psoriasis, the cumulative probability of drug survival for both IL-17 and IL-23 inhibitors was higher than 75% at 24 months, and IL-23 inhibitors had the highest drug survival rates during the entire study period110

Risankizumab and guselkumab demonstrated overall cumulative probabilities of ≥90%, and the likelihood of discontinuation was lower for isankizumab, guselkumab and ixekizumab, compared with secukinumab110. Predictors for drug discontinuation included the type of biologic, prior exposure to biologic agents, higher baseline body mass index (BMI) and PASI values, and absence of family history of psoriasis110.

However, it has been suggested that drug survival may not be sufficiently captured if other indicators of relative treatment failure (e.g. dose escalation) are not reflected84. An analysis of the Danish DERMBIO registry included dose escalation or drug discontinuation in their measure of biologic drug survival in patients with psoriasis during the first 3 years of treatment84. Results indicated 84:

  • secukinumab was associated with the lowest risk of the composite endpoint of either off-label dose escalation or discontinuation, compared with ustekinumab and adalimumab
  • a lower risk of this composite endpoint in biologic-naive versus biologic-experienced patients

Read about the place in therapy of psoriasis treatments

Explore effective treatment selection in psoriasis

Refresh your knowledge on European and US guidelines for psoriasis

References

  1. Nograles KE, Davidovici B, Krueger JG. New Insights in the Immunologic Basis of Psoriasis. Semin Cutan Med Surg. 2010;29(1):3–9.
  2. Nast A, Boehncke WH, Mrowietz U, Ockenfels HM, Philipp S, Reich K, et al. S3 - Guidelines on the treatment of psoriasis vulgaris (English version). Update. JDDG - Journal of the German Society of Dermatology. 2012;10(SUPPL.2):S1-s95.
  3. Boehncke WH, Boehncke S. More than skin-deep: The many dimensions of the psoriatic disease. Swiss Medical Weekly. 2014;144. doi:10.4414/smw.2014.13968.
  4. Nast A, Smith C, Spuls PI, Avila Valle G, Bata-Csörgö Z, Boonen H, et al. EuroGuiDerm Guideline on the systemic treatment of Psoriasis vulgaris – Part 1: treatment and monitoring recommendations. Journal of the European Academy of Dermatology and Venereology. 2020;34(11):2461–2498.
  5. Nast A, Smith C, Spuls PI, Avila Valle G, Bata-Csörgö Z, Boonen H, et al. EuroGuiDerm Guideline on the systemic treatment of Psoriasis vulgaris – Part 2: specific clinical and comorbid situations. Journal of the European Academy of Dermatology and Venereology. 2021;35(2):281–317.
  6. Elmets CA, Korman NJ, Prater EF, Wong EB, Rupani RN, Kivelevitch D, et al. Joint AAD–NPF Guidelines of care for the management and treatment of psoriasis with topical therapy and alternative medicine modalities for psoriasis severity measures. J Am Acad Dermatol. 2021;84(2):432–470.
  7. Menter A, Strober BE, Kaplan DH, Kivelevitch D, Prater EF, Stoff B, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with biologics. J Am Acad Dermatol. 2019;80(4):1029–1072.
  8. Menter A, Gelfand JM, Connor C, Armstrong AW, Cordoro KM, Davis DMR, et al. Joint American Academy of Dermatology–National Psoriasis Foundation guidelines of care for the management of psoriasis with systemic nonbiologic therapies. J Am Acad Dermatol. 2020;82(6):1445–1486.
  9. Reich K, Mrowietz U. Therapieziele bei der behandlung der psoriasis. JDDG - Journal of the German Society of Dermatology. 2007;5(7):566–574.
  10. Blome C, Gosau R, Radtke MA, Reich K, Rustenbach SJ, Spehr C, et al. Patient-relevant treatment goals in psoriasis. Arch Dermatol Res. 2016;308(2):69–78.
  11. Strober BE, van der Walt JM, Armstrong AW, Bourcier M, Carvalho AVE, Chouela E, et al. Clinical Goals and Barriers to Effective Psoriasis Care. Dermatol Ther (Heidelb). 2019;9(1):5–18.
  12. Papp K, Menter A, Poulin Y, Gu Y, Sasso EH. Long-term outcomes of interruption and retreatment vs. continuous therapy with adalimumab for psoriasis: Subanalysis of REVEAL and the open-label extension study. Journal of the European Academy of Dermatology and Venereology. 2013;27(5):634–642.
  13. Poulin Y, Sheth P, Gu Y. Psoriasis patients required to discontinue adalimumab therapy have worsening in their quality of life out of proportion to worsening in the objective signs of disease: Subanalysis of REVEAL. J Am Acad Dermatol. 2012;66(4):AB201.
  14. Okubo Y, Tsuruta D, Tang AC, Inoue S, Torisu-Itakura H, Hanada T, et al. Analysis of treatment goal alignment between Japanese psoriasis patients and their paired treating physicians. Journal of the European Academy of Dermatology and Venereology. 2018;32(4):606–614.
  15. Pintaudi B, Lucisano G, Gentile S, Bulotta A, Skovlund SE, Vespasiani G, et al. Correlates of diabetes-related distress in type 2 diabetes: Findings from the benchmarking network for clinical and humanistic outcomes in diabetes (BENCH-D) study. J Psychosom Res. 2015;79(5):348–354.
  16. Topp CW, Østergaard SD, Søndergaard S, Bech P. The WHO-5 Well-Being Index: A Systematic Review of the Literature. Psychother Psychosom. 2015;84(3):167–176.
  17. Mrowietz U, Sommer R, Reguiai Z, Gerdes S, Dauden E, Weger W, et al. Patient-reported well-being in value-based care using tildrakizumab in a real-world setting: 28-week interim data of the phase IV POSTIVE study. In: Poster presentation at the 25th World Congress of Dermatology 2023, July 3–8. 2023.
  18. Eurofound. European Quality of Life Survey - Data visualisation: WHO-5 mental well-being scale. European Quality of Life Survey 2016. 2016. https://www.eurofound.europa.eu/data/european-quality-of-life-survey?locale=EN&dataSource=EQLS2017NC&media=png&width=740&question=Y16_WHO5_MWIndex&plot=heatMap&countryGroup=linear&subset=Y16_Activity_status&subsetValue=All&answer=Mean. Accessed 4 July 2023.
  19. Omani-Samani R, Maroufizadeh S, Almasi-Hashiani A, Sepidarkish M, Amini P. The WHO-5 Well-Being Index: A Validation Study in People with Infertility. Iran J Public Health. 2019;48(11):2058–2064.
  20. Strober B, Papp KA, Lebwohl M, Reich K, Paul C, Blauvelt A, et al. Clinical meaningfulness of complete skin clearance in psoriasis. J Am Acad Dermatol. 2016;75(1):77-82.e7.
  21. Eghlileb AM, Davies EEG, Finlay AY. Psoriasis has a major secondary impact on the lives of family members and partners. British Journal of Dermatology. 2007;156(6):1245–1250.
  22. Armstrong A, Jarvis S, Boehncke WH, Rajagopalan M, Fernández-Peñas P, Romiti R, et al. Patient perceptions of clear/almost clear skin in moderate-to-severe plaque psoriasis: results of the Clear About Psoriasis worldwide survey. Journal of the European Academy of Dermatology and Venereology. 2018;32(12):2200–2207.
  23. Nestle FO, Kaplan DH, Barker J. Mechanisms of disease: Psoriasis. N Engl J Med. 2009;361(5):496–509.
  24. Boehncke WH, Schön MP. Psoriasis. The Lancet. 2015;386(9997):983–994.
  25. Palfreeman AC, McNamee KE, McCann FE. New developments in the management of psoriasis and psoriatic arthritis: A focus on apremilast. Drug Des Devel Ther. 2013;7:201–210.
  26. Sabat R, Philipp S, Höflich C, Kreutzer S, Wallace E, Asadullah K, et al. Immunopathogenesis of psoriasis. Exp Dermatol. 2007;16(10):779–798.
  27. Diani M, Altomare G, Reali E. T cell responses in psoriasis and psoriatic arthritis. Autoimmunity Reviews. 2015;14(4):286–292.
  28. Becher B, Pantelyushin S. Interleukin-17-producing γδ T cells go under the skin? Nat Med. 2012;18(12):1748–1750.
  29. Büchau AS, Gallo RL. Innate immunity and antimicrobial defense systems in psoriasis. Clinics in Dermatology. 2007;25(6):616–624.
  30. Chandrakumar SF, Yeung J. Interleukin-17 antagonists in the treatment of psoriasis. J Cutan Med Surg. 2015;19(2):109–114.
  31. Sugiyama H, Gyulai R, Toichi E, Garaczi E, Shimada S, Stevens SR, et al. Dysfunctional Blood and Target Tissue CD4 + CD25 high Regulatory T Cells in Psoriasis: Mechanism Underlying Unrestrained Pathogenic Effector T Cell Proliferation . The Journal of Immunology. 2005;174(1):164–173.
  32. Goodman WA, Levine AD, Massari J V., Sugiyama H, McCormick TS, Cooper KD. IL-6 Signaling in Psoriasis Prevents Immune Suppression by Regulatory T Cells. The Journal of Immunology. 2009;183(5):3170–3176.
  33. Brotas AM, Cunha JMT, Lago EHJ, Machado CCN, Carneiro SC da S. Fator de necrose tumoral-alfa e a rede de citocinas na psoriase. An Bras Dermatol. 2012;87(5):673–683.
  34. Ferreira VL, Borba, HL, de F. Bonetti, AF, Leonart, LP, Pontarolo R. Cytokines and Interferons: Types and Functions. In: Autoantibodies and Cytokines. 2019. IntechOpen: 65–87.
  35. Baliwag J, Barnes DH, Johnston A. Cytokines in psoriasis. 2015. Academic Press.
  36. Lowes MA, Suárez-Fariñas M, Krueger JG. Immunology of psoriasis. Annu Rev Immunol. 2014;32:227–255.
  37. Brunner PM, Koszik F, Reininger B, Kalb ML, Bauer W, Stingl G. Infliximab induces downregulation of the IL-12/IL-23 axis in 6-sulfo-LacNac (slan)+ dendritic cells and macrophages. Journal of Allergy and Clinical Immunology. 2013;132(5):1184–93.
  38. Soler DC, McCormick TS. The dark side of regulatory T cells in psoriasis. Journal of Investigative Dermatology. 2011;131(9):1785–1786.
  39. Amatya N, Garg A V., Gaffen SL. IL-17 Signaling: The Yin and the Yang. Trends Immunol. 2017;38(5):310–322.
  40. Lønnberg AS, Zachariae C, Skov L. Targeting of interleukin-17 in the treatment of psoriasis. Clin Cosmet Investig Dermatol. 2014;7:251–259.
  41. von Stebut E, Boehncke W-H, Ghoreschi K, Gori T, Kaya Z, Thaci D, et al. IL-17A in Psoriasis and Beyond: Cardiovascular and Metabolic Implications. Front Immunol. 2020;10. https://www.frontiersin.org/articles/10.3389/fimmu.2019.03096.
  42. Galluzzo M, D’Adamio S, Massaro A, Piccolo A, Bianchi L, Talamonti M. Spotlight on brodalumab in the treatment of plaque psoriasis: The evidence to date. Clin Cosmet Investig Dermatol. 2019;12:311–321.
  43. Di Meglio P, Nestle FO. The role of IL-23 in the immunopathogenesis of psoriasis. F1000 Biol Rep. 2010;2(1):40.
  44. Girolomoni G, Strohal R, Puig L, Bachelez H, Barker J, Boehncke WH, et al. The role of IL-23 and the IL-23/TH17 immune axis in the pathogenesis and treatment of psoriasis. Journal of the European Academy of Dermatology and Venereology. 2017;31(10):1616–1626.
  45. Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). The Lancet. 2008;371(9625):1665–1674.
  46. Langley RG, Lebwohl M, Krueger GG, Szapary PO, Wasfi Y, Chan D, et al. Long-term efficacy and safety of ustekinumab, with and without dosing adjustment, in patients with moderate-to-severe psoriasis: Results from the PHOENIX 2 study through 5 years of follow-up. British Journal of Dermatology. 2015;172(5):1371–1383.
  47. Gordon KB, Duffin KC, Bissonnette R, Prinz JC, Wasfi Y, Li S, et al. A Phase 2 Trial of Guselkumab versus Adalimumab for Plaque Psoriasis. New England Journal of Medicine. 2015;373(2):136–144.
  48. Blauvelt A, Papp KA, Griffiths CEM, Randazzo B, Wasfi Y, Shen YK, et al. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the continuous treatment of patients with moderate to severe psoriasis: Results from the phase III, double-blinded, placebo- and active comparator–. J Am Acad Dermatol. 2017;76(3):405–417.
  49. Reich K, Papp KA, Blauvelt A, Tyring SK, Sinclair R, Thaçi D, et al. Tildrakizumab versus placebo or etanercept for chronic plaque psoriasis (reSURFACE 1 and reSURFACE 2): results from two randomised controlled, phase 3 trials. The Lancet. 2017;390(10091):276–288.
  50. Gómez-García F, Ruano J, Gay-Mimbrera J, Aguilar-Luque M, Sanz-Cabanillas JL, Hernández Romero JL, et al. A Scoping Review Protocol to Explore the Use of Interleukin-1-Targeting Drugs for the Treatment of Dermatological Diseases: Indications, Mechanism of Action, Efficacy, and Safety. Dermatol Ther (Heidelb). 2018;8(2):195–202.
  51. Cai Y, Xue F, Quan C, Qu M, Liu N, Zhang Y, et al. A Critical Role of the IL-1β–IL-1R Signaling Pathway in Skin Inflammation and Psoriasis Pathogenesis. Journal of Investigative Dermatology. 2019;139(1):146–156.
  52. Sachen KL, Arnold Greving CN, Towne JE. Role of IL-36 cytokines in psoriasis and other inflammatory skin conditions. Cytokine. 2022;156:155897.
  53. Sugiura K. Role of Interleukin 36 in Generalised Pustular Psoriasis and Beyond. Dermatol Ther (Heidelb). 2022;12(2):315–328.
  54. Boutet M-A, Bart G, Penhoat M, Amiaud J, Brulin B, Charrier C, et al. Distinct expression of interleukin (IL)-36α, β and γ, their antagonist IL-36Ra and IL-38 in psoriasis, rheumatoid arthritis and Crohn’s disease. Clin Exp Immunol. 2016;184(2):159–173.
  55. Hao JQ. Targeting interleukin-22 in psoriasis. Inflammation. 2014;37(1):94–99.
  56. Chiricozzi A, Romanelli P, Volpe E, Borsellino G, Romanelli M. Scanning the immunopathogenesis of psoriasis. International Journal of Molecular Sciences. 2018;19(1):179.
  57. Wawrzycki B, Pietrzak A, Grywalska E, Krasowska D, Chodorowska G, Roliński J. Interleukin-22 and Its Correlation with Disease Activity in Plaque Psoriasis. Arch Immunol Ther Exp (Warsz). 2019;67(2):103–108.
  58. Reich K, Armstrong AW, Foley P, Song M, Wasfi Y, Randazzo B, et al. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the treatment of patients with moderate to severe psoriasis with randomized withdrawal and retreatment: Results from the phase III, double-blind, p. J Am Acad Dermatol. 2017;76(3):418–431.
  59. Steffen S, Abraham S, Herbig M, Schmidt F, Blau K, Meisterfeld S, et al. Toll-Like Receptor-Mediated Upregulation of CXCL16 in Psoriasis Orchestrates Neutrophil Activation. Journal of Investigative Dermatology. 2018;138(2):344–354.
  60. Kim WB, Jerome D, Yeung J. Diagnosis and management of psoriasis. Canadian Family Physician. 2017;63(4):278.
  61. Feldman SR, Goffe B, Rice G, Mitchell M, Kaur M, Robertson D, et al. The challenge of managing psoriasis: Unmet medical needs and stakeholder perspectives. Am Health Drug Benefits. 2016;9(9):504–512.
  62. Nast A, Gisondi P, Ormerod AD, Saiag P, Smith C, Spuls PI, et al. European S3-Guidelines on the systemic treatment of psoriasis vulgaris - Update 2015 - Short version - EDF in cooperation with EADV and IPC. Journal of the European Academy of Dermatology and Venereology. 2015;29(12):2277–2294.
  63. Mrowietz U, Kragballe K, Reich K, Spuls P, Griffiths CEM, Nast A, et al. Definition of treatment goals for moderate to severe psoriasis: A European consensus. Arch Dermatol Res. 2011;303(1):1–10.
  64. Pathirana D, Ormerod AD, Saiag P, Smith C, Spuls PI, Nast A, et al. European S3-guidelines on the systemic treatment of psoriasis vulgaris. Journal of the European Academy of Dermatology and Venereology. 2009;23(SUPPL. 2):1–70.
  65. Mrowietz U, Barker J, Boehncke WH, Iversen L, Kirby B, Naldi L, et al. Clinical use of dimethyl fumarate in moderate-to-severe plaque-type psoriasis: a European expert consensus. Journal of the European Academy of Dermatology and Venereology. 2018;32:3–14.
  66. Otezla (apremilast). Summary of product characteristics. 2015. Last updated 2022. https://www.ema.europa.eu/documents/product-information/otezla-epar-product-information_en.pdf. Accessed 5 May 2023.
  67. Electronic Medicines Compendium. Acitretin (Neotigason). Summary of product characteristics. 2023. https://www.medicines.org.uk/emc/product/10248/smpc. Accessed 5 May 2023.
  68. Skilarence (dimethyl fumarate). Summary of product characteristics. 2017. Last updated 2022. https://www.ema.europa.eu/documents/product-information/skilarence-epar-product-information_en.pdf. Accessed 5 May 2023.
  69. Nordimet (methotrexate). Summary of product characteristics. 2016. Last updated 2023. https://www.ema.europa.eu/documents/product-information/nordimet-epar-product-information_en.pdf. Accessed 5 May 2023.
  70. Jylamvo (methotrexate). Summary of product characteristics. 2017. Last updated 2023. https://www.ema.europa.eu/documents/product-information/jylamvo-epar-product-information_en.pdf. Accessed 5 May 2023.
  71. Benjamin O, Goyal A, Lappin S. Disease Modifying Anti-Rheumatic Drugs (DMARD). 2022. Treasure Island (FL). StatPearls Publishing.
  72. Yamauchi PS, Rizk D, Kormeili T, Patnaik R, Lowe NJ. Current systemic therapies for psoriasis: where are we now? J Am Acad Dermatol. 2003;49(2):66–77.
  73. Electronic Medicines Compendium. Capimune. Summary of Product Characteristics. 2021. https://www.medicines.org.uk/emc/product/695/smpc. Accessed 4 May 2023.
  74. Menter A, Korman NJ, Elmets CA, Feldman SR, Gelfand JM, Gordon KB, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis. Section 3. Guidelines of care for the management and treatment of psoriasis with topical therapies. J Am Acad Dermatol. 2009;60(4):643–659.
  75. Rivera R, Vilarrasa E, Ribera M, Roe E, Kueder-Pajares T, Zayas AI, et al. Unmet needs in patients with moderate-to-severe plaque psoriasis treated with methotrexate in real world practice: FirST study. Journal of Dermatological Treatment. 2020. doi:10.1080/09546634.2020.1801977.
  76. Dommasch ED, Kim SC, Lee MP, Gagne JJ. Risk of Serious Infection in Patients Receiving Systemic Medications for the Treatment of Psoriasis. JAMA Dermatol. 2019;155(10):1142–1152.
  77. Reich K, Mrowietz U, Radtke MA, Thaci D, Rustenbach SJ, Spehr C, et al. Drug safety of systemic treatments for psoriasis: results from The German Psoriasis Registry PsoBest. Arch Dermatol Res. 2015;307(10):875–883.
  78. Reszke R, Szepietowski JC. A safety evaluation of dimethyl fumarate in moderate-to-severe psoriasis. Expert Opin Drug Saf. 2020;19(4):373–380.
  79. Mrowietz U, Szepietowski JC, Loewe R, van de Kerkhof P, Lamarca R, Ocker WG, et al. Efficacy and safety of LAS41008 (dimethyl fumarate) in adults with moderate-to-severe chronic plaque psoriasis: a randomized, double-blind, Fumaderm®- and placebo-controlled trial (BRIDGE). British Journal of Dermatology. 2017;176(3):615–623.
  80. Naldi L, Griffiths CEM. Traditional therapies in the management of moderate to severe chronic plaque psoriasis: An assessment of the benefits and risks. British Journal of Dermatology. 2005;152(4):597–615.
  81. Zito P, Mazzoni T. Acitretin. 2023. Treasure Island (FL). StatPearls Publishing https://www.ncbi.nlm.nih.gov/books/NBK519571. Accessed 5 May 2023.
  82. European Medicines Agency. Otezla. Summary of Product Characteristics. 2021. https://www.ema.europa.eu/en/documents/product-information/otezla-epar-product-information_en.pdf. Accessed 17 February 2021.
  83. Papp K, Reich K, Leonardi CL, Kircik L, Chimenti S, Langley RGB, et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: Results of a phase III, randomized, controlled trial (Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J Am Acad Dermatol. 2015;73(1):37–49.
  84. Paul C, Cather J, Gooderham M, Poulin Y, Mrowietz U, Ferrandiz C, et al. Efficacy and safety of apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate-to-severe plaque psoriasis over 52 weeks: A phase III, randomized controlled trial (ESTEEM 2). British Journal of Dermatology. 2015;173(6):1387–1399.
  85. Bissonnette R, Pariser DM, Wasel NR, Goncalves J, Day RM, Chen R, et al. Apremilast, an oral phosphodiesterase-4 inhibitor, in the treatment of palmoplantar psoriasis: Results of a pooled analysis from phase II PSOR-005 and phase III Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis (ESTEEM) clinical . J Am Acad Dermatol. 2016;75(1):99–105.
  86. Rich P, Gooderham M, Bachelez H, Goncalves J, Day RM, Chen R, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with difficult-to-treat nail and scalp psoriasis: Results of 2 phase III randomized, controlled trials (ESTEEM 1 and ESTEEM 2). J Am Acad Dermatol. 2016;74(1):134–142.
  87. Thaçi D, Kimball A, Foley P, Poulin Y, Levi E, Chen R, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, improves patient-reported outcomes in the treatment of moderate to severe psoriasis: results of two phase III randomized, controlled trials. Journal of the European Academy of Dermatology and Venereology. 2017;31(3):498–506.
  88. Armstrong AW, Betts KA, Sundaram M, Thomason D, Signorovitch JE. Comparative efficacy and incremental cost per responder of methotrexate versus apremilast for methotrexate-naïve patients with psoriasis. J Am Acad Dermatol. 2016;75(4):740–746.
  89. Crowley J, Thaçi D, Joly P, Peris K, Papp KA, Goncalves J, et al. Long-term safety and tolerability of apremilast in patients with psoriasis: Pooled safety analysis for ≥156 weeks from 2 phase 3, randomized, controlled trials (ESTEEM 1 and 2). J Am Acad Dermatol. 2017;77(2):310-317.e1.
  90. Gisondi P, Del Giglio M, Girolomoni G. Treatment approaches tomoderate to severe psoriasis. Int J Mol Sci. 2017;18(11):2427.
  91. Cosentyx (secukinumab) EPAR. European Medicines Agency. 2023. https://www.ema.europa.eu/en/medicines/human/EPAR/cosentyx. Accessed 3 July 2023.
  92. Tremfya (guselkumab). Summary of product characteristics. 2017. Last updated 2022. https://www.ema.europa.eu/documents/product-information/tremfya-epar-product-information_en.pdf. Accessed 3 July 2023.
  93. Skyrizi (risankizumab). Summary of product characteristics. 2019. Last updated 2023. https://www.ema.europa.eu/documents/product-information/skyrizi-epar-product-information_en.pdf. Accessed 3 July 2023.
  94. Ilumetri (tildrakizumab). Summary of product characteristics. 2018. Last updated 2023. https://www.ema.europa.eu/documents/product-information/ilumetri-epar-product-information_en.pdf. Accessed 3 July 2023.
  95. Cosentyx (secukinumab). Summary of product characteristics. 2015. Last updated 2023. https://www.ema.europa.eu/documents/product-information/cosentyx-epar-product-information_en.pdf. Accessed 3 July 2023.
  96. Taltz (ixekizumab). Summary of product characteristics. 2016. Last updated 2023. https://www.ema.europa.eu/documents/product-information/taltz-epar-product-information_en.pdf. Accessed 3 July 2023.
  97. Sotyktu (deucravacitinib). Summary of product characteristics. 2023. https://www.ema.europa.eu/documents/product-information/sotyktu-epar-product-information_en.pdf. Accessed 3 July 2023.
  98. Spevigo (spesolimab). Summary of product characteristics. 2023. https://www.ema.europa.eu/documents/product-information/spevigo-epar-product-information_en.pdf. Accessed 3 July 2023.
  99. Kyntheum (brodalumab). Summary of product characteristics. 2017. Last updated 2023. https://www.ema.europa.eu/documents/product-information/kyntheum-epar-product-information_en.pdf. Accessed 3 July 2023.
  100. Benepali (etanercept). Summary of product characteristics. 2016. Last updated 2023. https://www.ema.europa.eu/documents/product-information/benepali-epar-product-information_en.pdf. Accessed 3 July 2023.
  101. Remsima (infliximab). Summary of product characteristics. 2013. Last updated 2023. https://www.ema.europa.eu/documents/product-information/remsima-epar-product-information_en.pdf. Accessed 3 July 2023.
  102. European Medicines Agency. Stelara (ustekinumab) EPAR. Last updated 2023. https://www.ema.europa.eu/en/medicines/human/EPAR/stelara. Accessed 3 July 2023.
  103. Cimzia (certolizumab pegol). Summary of product characteristics. 2010. Last updated 2023. https://www.ema.europa.eu/documents/product-information/cimzia-epar-product-information_en.pdf. Accessed 3 July 2023.
  104. Hukyndra (adalimumab). Summary of product characteristics. 2022. Last updated 2023. https://www.ema.europa.eu/documents/product-information/hukyndra-epar-product-information_en.pdf. Accessed 3 July 2023.
  105. Bimzelx (bimekizumab). Summary of product characteristics. 2021. Last updated 2022. https://www.ema.europa.eu/documents/product-information/bimzelx-epar-product-information_en.pdf. Accessed 3 July 2023.
  106. Menter A, Tyring SK, Gordon K, Kimball AB, Leonardi CL, Langley RG, et al. Adalimumab therapy for moderate to severe psoriasis: A randomized, controlled phase III trial. J Am Acad Dermatol. 2008;58(1):106–115.
  107. Papp KA, Yang M, Sundaram M, Jarvis J, Betts KA, Bao Y, et al. Comparison of Adalimumab and Etanercept for the Treatment of Moderate to Severe Psoriasis: An Indirect Comparison Using Individual Patient Data from Randomized Trials. Value in Health. 2018;21(1):1–8.
  108. Papp KA, Armstrong AW, Reich K, Karunaratne M, Valdecantos W. Adalimumab Efficacy in Patients with Psoriasis Who Received or Did Not Respond to Prior Systemic Therapy: A Pooled Post Hoc Analysis of Results from Three Double-Blind, Placebo-Controlled Clinical Trials. Am J Clin Dermatol. 2016;17(1):79–86.
  109. Menter A, Thaçi D, Wu JJ, Abramovits W, Kerdel F, Arikan D, et al. Long-Term Safety and Effectiveness of Adalimumab for Moderate to Severe Psoriasis: Results from 7-Year Interim Analysis of the ESPRIT Registry. Dermatol Ther (Heidelb). 2017;7(3):365–381.
  110. Wu JJ, Abramovits W, Valdecantos WC, Servin OR, Bereswill M, Arikan D, et al. 14030 Ten-year interim results from the ESPRIT registry: Real-world safety, effectiveness, and patient-reported outcomes of adalimumab for moderate to severe psoriasis. J Am Acad Dermatol. 2020;83(6):AB17.
  111. Gisondi P, Geat D, Pizzolato M, Girolomoni G. State of the art and pharmacological pipeline of biologics for chronic plaque psoriasis. Curr Opin Pharmacol. 2019;46:90–99.
  112. ClinicalTrials.gov. NCT02326272 - A Study to Evaluate the Efficacy and Safety of Two Dose Levels of Certolizumab Pegol (CZP) in Subjects With Plaque Psoriasis (PSO). 2018. https://clinicaltrials.gov/ct2/show/NCT02326272. Accessed 24 February 2021.
  113. Cochrane Central Register of Controlled Trials (CENTRAL). Efficacy and Safety Study of Sirukumab in Subjects With Polymyalgia Rheumatica. 2016.
  114. Rutkowski D, Chinoy H, Warren RB. The Potential Benefits of Certolizumab Pegol in Patients with Concurrent Psoriatic Arthritis and Chronic Plaque Psoriasis: A Case Series and Review of the Literature. Dermatol Ther (Heidelb). 2019;9(2):373–381.
  115. Gottlieb AB, Blauvelt A, Thaçi D, Leonardi CL, Poulin Y, Drew J, et al. Certolizumab pegol for the treatment of chronic plaque psoriasis: Results through 48 weeks from 2 phase 3, multicenter, randomized, double-blinded, placebo-controlled studies (CIMPASI-1 and CIMPASI-2). J Am Acad Dermatol. 2018;79(2):302-314.e6.
  116. Lebwohl M, Blauvelt A, Paul C, Sofen H, Węgłowska J, Piguet V, et al. Certolizumab pegol for the treatment of chronic plaque psoriasis: Results through 48 weeks of a phase 3, multicenter, randomized, double-blind, etanercept- and placebo-controlled study (CIMPACT). J Am Acad Dermatol. 2018;79(2):266-276.e5.
  117. Curtis JR, Mariette X, Gaujoux-Viala C, Blauvelt A, Kvien TK, Sandborn WJ, et al. Long-term safety of certolizumab pegol in rheumatoid arthritis, axial spondyloarthritis, psoriatic arthritis, psoriasis and Crohn’s disease: A pooled analysis of 11 317 patients across clinical trials. RMD Open. 2019;5(1). doi:10.1136/rmdopen-2019-000942.
  118. Blauvelt A, Paul C, van de Kerkhof P, Warren RB, Gottlieb AB, Langley RG, et al. Long-term safety of certolizumab pegol in plaque psoriasis: pooled analysis over 3 years from three phase III, randomized, placebo-controlled studies. British Journal of Dermatology. 2021;184(4):640–651.
  119. Mariette X, Förger F, Abraham B, Flynn AD, Moltó A, Flipo RM, et al. Lack of placental transfer of certolizumab pegol during pregnancy: Results from CRIB, a prospective, postmarketing, pharmacokinetic study. Ann Rheum Dis. 2018;77(2):228–233.
  120. Clowse ME, Förger F, Hwang C, Thorp J, Dolhain RJ, Van Tubergen A, et al. Minimal to no transfer of certolizumab pegol into breast milk: Results from CRADLE, a prospective, postmarketing, multicentre, pharmacokinetic study. Ann Rheum Dis. 2017;76(11):1890–1896.
  121. Lee JY, Kang S, Park JS, Jo SJ. Prevalence of psoriasis in Korea: A population-based epidemiological study using the Korean national health insurance database. Ann Dermatol. 2017;29(6):761–767.
  122. Luger T, Schopf RE, Schwanke A, Langhammer S, Meng T, Löschmann PA. An observational study to evaluate the long-term outcomes of treatment with etanercept in patients with plaque-type psoriasis. Journal of the European Academy of Dermatology and Venereology. 2016;30(10):1730–1741.
  123. Leonardi CL, Powers JL, Matheson RT, Goffe BS, Zitnik R, Wang A, et al. Etanercept as Monotherapy in Patients with Psoriasis. New England Journal of Medicine. 2003;349(21):2014–2022.
  124. Papp KA. The long-term efficacy and safety of new biological therapies for psoriasis. Arch Dermatol Res. 2006;298(1):7–15.
  125. Kimball AB, Rothman KJ, Kricorian G, Pariser D, Yamauchi PS, Menter A, et al. OBSERVE-5: Observational postmarketing safety surveillance registry of etanercept for the treatment of psoriasis final 5-year results. J Am Acad Dermatol. 2015;72(1):115–122.
  126. Subedi S, Gong Y, Chen Y, Shi Y. Infliximab and biosimilar infliximab in psoriasis: Efficacy, loss of efficacy, and adverse events. Drug Des Devel Ther. 2019;13:2491–2502.
  127. Reich K, Wozel G, Zheng H, Van Hoogstraten HJF, Flint L, Barker J. Efficacy and safety of infliximab as continuous or intermittent therapy in patients with moderate-to-severe plaque psoriasis: Results of a randomized, long-term extension trial (RESTORE2). British Journal of Dermatology. 2013;168(6):1325–1334.
  128. Shear NH, Hartmann M, Toledo-Bahena M, Katsambas A, Connors L, Chang Q, et al. Long-term efficacy and safety of infliximab maintenance therapy in patients with plaque-type psoriasis in real-world practice. British Journal of Dermatology. 2014;171(3):631–641.
  129. Kim WB, Marinas JEC, Qiang J, Shahbaz A, Greaves S, Yeung J. Adverse events resulting in withdrawal of biologic therapy for psoriasis in real-world clinical practice: A Canadian multicenter retrospective study. J Am Acad Dermatol. 2015;73(2):237–241.
  130. Landells I, Marano C, Hsu MC, Li S, Zhu Y, Eichenfield LF, et al. Ustekinumab in adolescent patients age 12 to 17 years with moderate-to-severe plaque psoriasis: Results of the randomized phase 3 CADMUS study. J Am Acad Dermatol. 2015;73(4):594–603.
  131. Papp K, Gottlieb AB, Naldi L, Pariser D, Ho V, Goyal K, et al. Safety surveillance for ustekinumab and other psoriasis treatments from the psoriasis longitudinal assessment and (PSOLAR). Journal of Drugs in Dermatology. 2015;14(7):706–714.
  132. Strober BE, Bissonnette R, Fiorentino D, Kimball AB, Naldi L, Shear NH, et al. Comparative effectiveness of biologic agents for the treatment of psoriasis in a real-world setting: Results from a large, prospective, observational study (Psoriasis Longitudinal Assessment and Registry [PSOLAR]). J Am Acad Dermatol. 2016;74(5):851-861.e4.
  133. Menter A, Papp KA, Gooderham M, Pariser DM, Augustin M, Kerdel FA, et al. Drug survival of biologic therapy in a large, disease-based registry of patients with psoriasis: results from the Psoriasis Longitudinal Assessment and Registry (PSOLAR). Journal of the European Academy of Dermatology and Venereology. 2016;30(7):1148–1158.
  134. Egeberg A, Rosenø NAL, Aagaard D, Lørup EH, Nielsen ML, Nymand L, et al. Drug survival of biologics and novel immunomodulators for rheumatoid arthritis, axial spondyloarthritis, psoriatic arthritis, and psoriasis - A nationwide cohort study from the DANBIO and DERMBIO registries. Semin Arthritis Rheum. 2022;53:151979.
  135. Augustin M, Abeysinghe S, Mallya U, Qureshi A, Roskell N, McBride D, et al. Secukinumab treatment of plaque psoriasis shows early improvement in DLQI response - Results of a phase II regimen-finding trial. Journal of the European Academy of Dermatology and Venereology. 2016;30(4):645–649.
  136. Zweegers J, Groenewoud JMM, van den Reek JMPA, Otero ME, van de Kerkhof PCM, Driessen RJB, et al. Comparison of the 1- and 5-year effectiveness of adalimumab, etanercept and ustekinumab in patients with psoriasis in daily clinical practice: results from the prospective BioCAPTURE registry. British Journal of Dermatology. 2017;176(4):1001–1009.
  137. Bagel J, Blauvelt A, Nia J, Hashim P, Patekar M, de Vera A, et al. Secukinumab maintains superiority over ustekinumab in clearing skin and improving quality of life in patients with moderate to severe plaque psoriasis: 52-week results from a double-blind phase 3b trial (CLARITY). Journal of the European Academy of Dermatology and Venereology. 2021;35(1):135–142.
  138. Reich K, Papp KA, Blauvelt A, Langley RG, Armstrong A, Warren RB, et al. Bimekizumab versus ustekinumab for the treatment of moderate to severe plaque psoriasis (BE VIVID): efficacy and safety from a 52-week, multicentre, double-blind, active comparator and placebo controlled phase 3 trial. Lancet. 2021;397(10273):487–498.
  139. Hawkes JE, Chan TC, Krueger JG. Psoriasis pathogenesis and the development of novel targeted immune therapies. Journal of Allergy and Clinical Immunology. 2017;140(3):645–653.
  140. Kim J, Krueger JG. Highly Effective New Treatments for Psoriasis Target the IL-23/Type 17 T Cell Autoimmune Axis. Annu Rev Med. 2017;68:255–269.
  141. Chan TC, Hawkes JE, Krueger JG. Interleukin 23 in the skin: role in psoriasis pathogenesis and selective interleukin 23 blockade as treatment. Ther Adv Chronic Dis. 2018;9(5):111–119.
  142. European Medicines Compendium. Tremfya. Summary of Product Characteristics. 2021. https://www.medicines.org.uk/emc/product/9587. Accessed 19 February 2021.
  143. European Medicines Compendium. Ozurdex. Summary of Prescribing Information. 2021. https://www.medicines.org.uk/emc/product/5654/smpc#gref. Accessed 17 February 2021.
  144. Sun Pharma. Sun Pharma announces receipt of European Commission approval for ILUMETRI® (tildrakizumab) by Almirall for treatment of moderate-to-severe chronic plaque psoriasis. 2018. www.sunpharma.com/Media/Press-Releases/Press Release European Commission Approval For ILUMETRI.pdf. Accessed 17 February 2021.
  145. European Medicines Compendium. Flixabi. Summary of Product Characteristics. 2021. https://www.medicines.org.uk/emc/product/7265/smpc. Accessed 5 January 2022.
  146. Nogueira M, Torres T. Guselkumab for the treatment of psoriasis – evidence to date. Drugs Context. 2019;8:212594.
  147. Pithadia DJ, Reynolds KA, Lee EB, Liao W, Wu JJ. Tildrakizumab in the treatment of psoriasis: latest evidence and place in therapy. Ther Adv Chronic Dis. 2019;10. doi:10.1177/2040622319865658.
  148. Reich K, Warren RB, Iversen L, Puig L, Pau-Charles I, Igarashi A, et al. Long-term efficacy and safety of tildrakizumab for moderate-to-severe psoriasis: pooled analyses of two randomized phase III clinical trials (reSURFACE 1 and reSURFACE 2) through 148 weeks. British Journal of Dermatology. 2020;182(3):605–617.
  149. Thaci D, Piaserico S, Warren RB, Gupta AK, Cantrell W, Draelos Z, et al. Five-year efficacy and safety of tildrakizumab in patients with moderate-to-severe psoriasis who respond at week 28: pooled analyses of two randomized phase III clinical trials (reSURFACE 1 and reSURFACE 2)*. British Journal of Dermatology. 2021;185(2):323–334.
  150. Drerup KA, Seemann C, Gerdes S, Mrowietz U. Effective and Safe Treatment of Psoriatic Disease with the Anti-IL-23p19 Biologic Tildrakizumab: Results of a Real-World Prospective Cohort Study in Nonselected Patients. Dermatology. 2021. doi:10.1159/000519924.
  151. Costanzo A, Llamas-Velasco M, Fabbrocini G, Cuccia A, Rivera-Diaz R, Gaarn Du Jardin K, et al. Tildrakizumab improves high burden skin symptoms, impaired sleep and quality of life of moderate-to-severe plaque psoriasis patients in conditions close to clinical practice. Journal of the European Academy of Dermatology and Venereology. 2023;n/a(n/a). doi:https://doi.org/10.1111/jdv.19229.
  152. Thaci D, Piaserico S, Warren RB, Gupta AK, Cantrell W, Draelos Z, et al. Five-year efficacy and safety of tildrakizumab in patients with moderate-to-severe psoriasis who respond at week 28: pooled analyses of two randomized phase III clinical trials (reSURFACE 1 and reSURFACE 2)*. British Journal of Dermatology. 2021;185(2):323–334.
  153. Banaszczyk K. Risankizumab in the treatment of psoriasis - Literature review. Reumatologia. 2019;57(3):158–162.
  154. Machado Á, Torres T. Spotlight on risankizumab and its potential in the treatment of plaque psoriasis: evidence to date. Psoriasis: Targets and Therapy. 2018;Volume 8:83–92.
  155. Strober B, Menter A, Leonardi C, Gordon K, Lambert J, Puig L, et al. Efficacy of risankizumab in patients with moderate-to-severe plaque psoriasis by baseline demographics, disease characteristics and prior biologic therapy: an integrated analysis of the phase III UltIMMa-1 and UltIMMa-2 studies. Journal of the European Academy of Dermatology and Venereology. 2020;34(12):2830–2838.
  156. Papp KA, Lebwohl MG, Puig L, Ohtsuki M, Beissert S, Zeng J, et al. Long-term efficacy and safety of risankizumab for the treatment of moderate-to-severe plaque psoriasis: interim analysis of the LIMMitless open-label extension trial beyond 3 years of follow-up. Br J Dermatol. 2021;185(6):1135–1145.
  157. Gordon KB, Strober B, Lebwohl M, Augustin M, Blauvelt A, Poulin Y, et al. Efficacy and safety of risankizumab in moderate-to-severe plaque psoriasis (UltIMMa-1 and UltIMMa-2): results from two double-blind, randomised, placebo-controlled and ustekinumab-controlled phase 3 trials. The Lancet. 2018;392(10148):650–661.
  158. Yamauchi PS, Bagel J. Next-generation biologics in the management of plaque psoriasis: A literature review of IL-17 inhibition. Journal of Drugs in Dermatology. 2015;14(3):244–250.
  159. Koppu S, Singh R, Kaur K, Feldman SR. Review of bimekizumab in the treatment of psoriasis. Hum Vaccin Immunother. 2022;18(6):2119767.
  160. Reich K, Warren RB, Lebwohl M, Gooderham M, Strober B, Langley RG, et al. Bimekizumab versus Secukinumab in Plaque Psoriasis. New England Journal of Medicine. 2021;385(2):142–152.
  161. Warren RB, Blauvelt A, Bagel J, Papp KA, Yamauchi P, Armstrong A, et al. Bimekizumab versus Adalimumab in Plaque Psoriasis. New England Journal of Medicine. 2021;385(2):130–141.
  162. Gordon KB, Foley P, Krueger JG, Pinter A, Reich K, Vender R, et al. Bimekizumab efficacy and safety in moderate to severe plaque psoriasis (BE READY): a multicentre, double-blind, placebo-controlled, randomised withdrawal phase 3 trial. The Lancet. 2021;397(10273):475–486.
  163. Reich K, Papp KA, Blauvelt A, Langley RG, Armstrong A, Warren RB, et al. Bimekizumab versus ustekinumab for the treatment of moderate to severe plaque psoriasis (BE VIVID): efficacy and safety from a 52-week, multicentre, double-blind, active comparator and placebo controlled phase 3 trial. The Lancet. 2021;397(10273):487–498.
  164. Foulkes AC, Warren RB. Brodalumab in psoriasis: Evidence to date and clinical potential. Drugs Context. 2019;8. doi:10.7573/dic.212570.
  165. Pinter A, Bonnekoh B, Hadshiew IM, Zimmer S. Brodalumab for the treatment of moderate-tosevere psoriasis: Case series and literature review. Clin Cosmet Investig Dermatol. 2019;12:509–517.
  166. Puig L, Lebwohl M, Bachelez H, Sobell J, Jacobson AA. Long-term efficacy and safety of brodalumab in the treatment of psoriasis: 120-week results from the randomized, double-blind, placebo- and active comparator–controlled phase 3 AMAGINE-2 trial. J Am Acad Dermatol. 2020;82(2):352–359.
  167. Bissonnette R, Luger T, Thaçi D, Toth D, Lacombe A, Xia S, et al. Secukinumab demonstrates high sustained efficacy and a favourable safety profile in patients with moderate-to-severe psoriasis through 5 years of treatment (SCULPTURE Extension Study). Journal of the European Academy of Dermatology and Venereology. 2018;32(9):1507–1514.
  168. Feldman SR, Gomez B, Meng X, Germino R. Secukinumab rapidly improves EQ-5D health status in patients with psoriasis: Pooled analysis from four phase 3 trials. Journal of Dermatological Treatment. 2021;32(7):709–715.
  169. Blauvelt A, Reich K, Tsai TF, Tyring S, Vanaclocha F, Kingo K, et al. Secukinumab is superior to ustekinumab in clearing skin of subjects with moderate-to-severe plaque psoriasis up to 1 year: Results from the CLEAR study. J Am Acad Dermatol. 2017;76(1):60-69.e9.
  170. Magnolo N, Kingo K, Laquer V, Browning J, Reich A, Szepietowski JC, et al. A phase 3 open-label, randomized multicenter study to evaluate efficacy and safety of secukinumab in pediatric patients with moderate to severe plaque psoriasis: 24-week results. J Am Acad Dermatol. 2022;86(1):122–130.
  171. Van De Kerkhof PCM, Griffiths CEM, Reich K, Leonardi CL, Blauvelt A, Tsai TF, et al. Secukinumab long-term safety experience: A pooled analysis of 10 phase II and III clinical studies in patients with moderate to severe plaque psoriasis. J Am Acad Dermatol. 2016;75(1):83-98.e4.
  172. Khattri S, Goldblum O, Solotkin K, Amir Y, Min MS, Ridenour T, et al. Early onset of clinical improvement with Ixekizumab in a Randomized, open-label study of patients with moderate-to-severe plaque psoriasis. Journal of Clinical and Aesthetic Dermatology. 2018;11(5):33–37.
  173. Sekhon S, Jeon C, Nakamura M, Yan D, Afifi L, Bhutani T, et al. Clinical utility of ixekizumab in the treatment of moderate-to-severe plaque psoriasis. Psoriasis: Targets and Therapy. 2017;7:65–72.
  174. Strober B, Sigurgeirsson B, Popp G, Sinclair R, Krell J, Stonkus S, et al. Secukinumab improves patient-reported psoriasis symptoms of itching, pain, and scaling: Results of two phase 3, randomized, placebo-controlled clinical trials. Int J Dermatol. 2016;55(4):401–407.
  175. Langley RG, Kimball AB, Nak H, Xu W, Pangallo B, Osuntokun OO, et al. Long-term safety profile of ixekizumab in patients with moderate-to-severe plaque psoriasis: an integrated analysis from 11 clinical trials. Journal of the European Academy of Dermatology and Venereology. 2019;33(2):333–339.
  176. Boehringer Ingelheim. European Commission approves SPEVIGO® (spesolimab) for generalized pustular psoriasis flares. Press release. 2022. https://www.boehringer-ingelheim.com/human-health/skin-diseases/gpp/european-commission-approves-spevigo-spesolimab-generalized. Accessed 10 May 2023.
  177. European Medicines Agency. Spevigo (spesolimab) EPAR. 2023. https://www.ema.europa.eu/en/medicines/human/EPAR/spevigo. Accessed 10 May 2023.
  178. Bachelez H, Choon S-E, Marrakchi S, Burden AD, Tsai T-F, Morita A, et al. Trial of Spesolimab for Generalized Pustular Psoriasis. New England Journal of Medicine. 2021;385(26):2431–2440.
  179. European Medicines Agency. Sotyktu (deucravacitinib) EPAR. 2023. https://www.ema.europa.eu/en/medicines/human/EPAR/sotyktu. Accessed 11 May 2023.
  180. Armstrong AW, Gooderham M, Warren RB, Papp KA, Strober B, Thaçi D, et al. Deucravacitinib versus placebo and apremilast in moderate to severe plaque psoriasis: Efficacy and safety results from the 52-week, randomized, double-blinded, placebo-controlled phase 3 POETYK PSO-1 trial. J Am Acad Dermatol. 2023;88(1):29–39.
  181. Strober B, Thaçi D, Sofen H, Kircik L, Gordon KB, Foley P, et al. Deucravacitinib versus placebo and apremilast in moderate to severe plaque psoriasis: Efficacy and safety results from the 52-week, randomized, double-blinded, phase 3 Program fOr Evaluation of TYK2 inhibitor psoriasis second trial. J Am Acad Dermatol. 2023;88(1):40–51.
  182. European Medicines Agency. Sotyktu (deucravacitinib) EPAR - Medicine overview. 2023 https://www.ema.europa.eu/documents/overview/sotyktu-epar-medicine-overview_en.pdf. Accessed 11 May 2023.
  183. Martin G. Novel Therapies in Plaque Psoriasis: A Review of Tyrosine Kinase 2 Inhibitors. Dermatol Ther (Heidelb). 2023;13(2):417–435.
  184. McInnes IB, Byers NL, Higgs RE, Lee J, Macias WL, Na S, et al. Comparison of baricitinib, upadacitinib, and tofacitinib mediated regulation of cytokine signaling in human leukocyte subpopulations. Arthritis Res Ther. 2019;21(1):183.
  185. Cohen S, Barer F, Itzhak I, Silverman MH, Fishman P. Inhibition of IL-17 and IL-23 in Human Keratinocytes by the A3 Adenosine Receptor Agonist Piclidenoson. J Immunol Res. 2018;2018:2310970.
  186. Clinicaltrials.gov. 2023. https://clinicaltrials.gov/. Accessed 9 May 2023.
  187. EU clinical trials register. 2023. https://www.clinicaltrialsregister.eu/ctr-search/search. Accessed 9 May 2023.
  188. Nast A, Altenburg A, Augustin M, Boehncke W-H, Härle P, Klaus J, et al. German S3-Guideline on the treatment of Psoriasis vulgaris, adapted from EuroGuiDerm – Part 2: Treatment monitoring and specific clinical or comorbid situations. JDDG: Journal der Deutschen Dermatologischen Gesellschaft. 2021;19(7):1092–1115.
  189. Nast A, Altenburg A, Augustin M, Boehncke W-H, Härle P, Klaus J, et al. German S3-Guideline on the treatment of Psoriasis vulgaris, adapted from EuroGuiDerm – Part 1: Treatment goals and treatment recommendations. JDDG: Journal der Deutschen Dermatologischen Gesellschaft. 2021;19(6):150–934.
  190. Lambert JLW, Segaert S, Ghislain PD, Hillary T, Nikkels A, Willaert F, et al. Practical recommendations for systemic treatment in psoriasis according to age, pregnancy, metabolic syndrome, mental health, psoriasis subtype and treatment history (BETA-PSO: Belgian Evidence-based Treatment Advice in Psoriasis; part 1). Journal of the European Academy of Dermatology and Venereology. 2020;34(8):1654–1665.
Welcome: