Maintenance of response with certolizumab pegol for the treatment of chronic plaque psoriasis: 48-week results from two ongoing Phase III, multicenter, randomized, placebo-controlled studies (CIMPASI-1 and CIMPASI-2)

Maintenance of response with certolizumab pegol for the treatment of chronic plaque psoriasis: 48-week results from two ongoing Phase III, multicenter, randomized, placebo-controlled studies (CIMPASI-1 and CIMPASI-2)

Presenters: Reich K1, Blauvelt A2, Thaçi D3, Leonardi C4, Poulin Y5, Burge D6, Peterson L7, Drew J6, Arendt C8, Gottlieb AB9

Affiliations: 1Dermatologikum Hamburg and SCIderm Research Institute, Hamburg, Germany; 2Oregon Medical Research Center, Portland, OR; 3University of Lübeck, Lübeck, Germany; 4Central Dermatology and Saint Louis University School of Medicine, St. Louis, MO; 5Centre de Recherche Dermatologique du Québec Métropolitain, Québec, Canada; 6Dermira, Inc., Menlo Park, CA; 7UCB BioSciences, Inc., Raleigh, NC; 8UCB Pharma, Brussels, Belgium; 9New York Medical College, Valhalla, NY

Introduction: Psoriasis affects approximately three percent of adults in the United States and approximately 2 to 6 percent in Europe, and most patients develop the disease in the third decade of life. Therapy for patients with chronic plaque psoriasis varies per the severity of the disease, with topical therapies and/or phototherapy used to treat limited or mild psoriasis, and photochemotherapy, cyclosporine, methotrexate, apremilast, or biologics (e.g., tumor necrosis factor (TNF) inhibitors,

anti?IL17s, and anti?IL12/23s) used to treat moderate-to-severe forms. Certolizumab pegol (CZP) is the only PEGylated, Fc-free, anti-TNF biologic currently under development for the treatment of moderate-to-severe chronic plaque psoriasis and has demonstrated positive results in previous psoriasis trials. CIMPASI-1 (NCT02326298) and CIMPASI-2 (NCT02326272) are ongoing Phase III trials designed to assess the efficacy and safety of CZP compared with placebo; results from the first 48 weeks of the two studies are presented here.

Methods: CIMPASI-1 and CIMPASI-2 are replicate, Phase III multicenter studies conducted in North America and Europe, consisting of randomized, double-blind, placebo-controlled periods for the first 48 weeks followed by 96 weeks of open-label observation. Patients were randomized 2:2:1 to CZP 400mg every two weeks (Q2W), CZP 200mg Q2W (following 400mg loading dose at Weeks 0, 2, 4), or placebo Q2W for 16 weeks. At Week 16, the following patients continued to receive treatment through Week 48: CZP 400mg Q2W- and CZP 200mg Q2W-treated Psoriasis Area and Severity Index (PASI) 50 responders (?50% reduction in PASI) continued to receive their initial blinded treatment; placebo-treated Week 16 PASI 75 responders (?75% reduction in PASI) continued blinded placebo treatment; PASI 50 to 75 responders (?50% but <75% reduction in PASI) received CZP 200mg Q2W (following 400mg loading dose at Weeks 16, 18, 20); PASI 50 nonresponders at Week 16 entered the Escape Arm and received unblinded CZP 400mg Q2W; PASI 50 nonresponders at Week 32, 40, or 48 were withdrawn from the study. Eligible patients were at least 18 years of age and had moderate-to-severe chronic plaque psoriasis for at least six months (PASI ?12, Body Surface Area (BSA) ?10%, Physician’s Global Assessment [PGA; 5-point scale] ?3). Patients had to be candidates for systemic psoriasis therapy, phototherapy, and/or photochemotherapy. Patients were excluded if they had previous treatment with CZP or more than two biologics (including anti-TNF); had history of primary failure to any biologic or secondary failure to more than one biologic; had erythrodermic, guttate, or generalized pustular psoriasis types; or had history of current, chronic, or recurrent viral, bacterial, or fungal infections. Coprimary endpoints were PASI 75 and PGA 0/1 (“clear” or “almost clear” with ?2-category improvement) at Week 16. Secondary endpoints reported here were PASI 90 at Week 16 and PASI 75 and PGA 0/1 at Week 48; PASI 90 at Week 48 was included as an additional endpoint. All efficacy analyses were performed on the Randomized Set (all randomized patients). Logistic regression models with factors of treatment group, region, and prior biologic exposure (yes/no) were used to analyze PASI 75, PGA 0/1, and PASI 90 responder rates (Week 16). The Markov chain Monte Carlo (MCMC) method for multiple imputation was used to account for missing data. Safety assessments were performed on the Safety Set, which included all randomized patients who received at least one dose of study medication.

Results: In both studies, at least 90 percent of patients in each treatment arm completed Week 16, and the highest Week 16 completion rates were in the CZP 400mg Q2W treatment group. Of those patients who entered the blinded Maintenance Period in CIMPASI-1/CIMPASI-2, 90.9 percent/88.4 percent of CZP 400mg Q2W patients and 95.9 percent/84.2 percent CZP 200mg Q2W patients completed Week 48. Baseline PASI and PGA scores were comparable across treatment groups for both studies. Roughly one-third of study participants reported prior biologic use, and the proportion across treatment arms was similar. At Week 16, responder rates were greater for CZP 400mg Q2W and CZP 200mg Q2W versus placebo for PASI 75 and PGA 0/1 (p<0.0001 for all). CZP 400mg Q2W and CZP 200mg Q2W PASI 75 responder rates were maintained to Week 48. PGA 0/1 responder rates were also maintained to Week 48 for patients who continued to receive either dose of CZP during the Maintenance Period. At Week 16, PASI 90 responder rates were greater for CZP 400mg Q2W and 200mg Q2W versus placebo (p<0.0001 for both). PASI 90 responder rates continued to improve beyond Week 16, and the difference between dose groups increased by Week 48. For CZP 400mg Q2W and CZP 200mg Q2W versus placebo, treatment-emergent adverse events (TEAE)/serious TEAE incidence rates per 100 patient?years from baseline to Week 16 were 375.9/19.0 and 292.3/6.9 versus 297.1/6.8 in CIMPASI-1, and 405.7/15.3 and 308.7/7.4 versus 388.9/0 in CIMPASI-2. For CZP 400mg Q2W and CZP 200mg Q2W, TEAE/serious TEAE incidence rates per 100 patient-years was lower from baseline to Week 48 compared with rates per 100 patient-years from baseline to Week 16 (257.6/10.4 and 218.3/5.3 in CIMPASI-1, and 277.5/7.5 and 236.0/9.7 in CIMPASI-2). One serious infection was reported in the CZP 400mg Q2W group in CIMPASI-1, and one in the

CZP 400mg Q2W group in CIMPASI-2. One death, due to motor vehicle accident, was reported in the

CZP 400mg Q2W group in CIMPASI?1. After 48 weeks of treatment, TEAEs occurring in over 10 percent of all CZP-treated patients were nasopharyngitis and upper respiratory tract infection.

Conclusion: Treatment with CZP 400mg Q2W or CZP 200mg Q2W for 16 weeks was associated with statistically significant, clinically meaningful improvements for all endpoints analyzed (PASI 75, PGA 0/1, and PASI 90) compared to placebo. Response rates were maintained at Week 48 with both CZP doses. For most measures, improvement at both Week 16 and Week 48 was numerically greatest in patients receiving CZP 400mg Q2W. TEAEs were consistent with the known safety profile of anti-TNF therapy, and no new safety signals were observed with CZP at any dose over 48 weeks.

Funding/Disclosures: This study and all costs associated with the development of this poster were funded by Dermira, Inc. Andrew Blauvelt and Diamant Thaçi receive consulting honoraria, are clinical investigators for and/or receive speaker’s fees from Dermira, Inc. Daniel Burge and Janice Drew are employees of Dermira, Inc. Alice B. Gottlieb holds consulting and/or advisory board agreements with Dermira, Inc.

Efficacy and safety of apremilast in systemic- and biologic-naive patients with moderate plaque psoriasis (52-week results of the UNVEIL study)

Efficacy and safety of apremilast in systemic- and biologic-naive patients with moderate plaque psoriasis (52-week results of the UNVEIL study)

Presenters: Strober B1, Forman S2, Bagel J3, Lebwohl M4, Stein Gold L5, Jackson JM6, Goncalves J7, Levi E7, Callis Duffin K8

Affiliations: 1University of Connecticut, Farmington, CT, and Probity Medical Research, Waterloo, ON, Canada; 2Forward Clinical Trials, Tampa, FL; 3Psoriasis Treatment Center of Central New Jersey, East Windsor, NJ; 4Icahn School of Medicine at Mount Sinai, New York, NY; 5Henry Ford Health System, West Bloomfield, MI; 6University of Louisville, Forefront Dermatology, Louisville, KY; 7Celgene Corporation, Summit, NJ; 8University of Utah, Salt Lake City, UT

Background/Objective: Patients with moderate plaque psoriasis (i.e., 5–10% psoriasis-involved Body Surface Area [BSA]) often receive no treatment or are undertreated with topical monotherapy. Apremilast, an oral, small-molecule phosphodiesterase 4 inhibitor, has been shown to be effective and has demonstrated acceptable tolerability in patients with moderate-to-severe psoriasis (BSA 10%) in the Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis (ESTEEM) Phase III clinical trial program. Evaluating Apremilast in a Phase IV Trial of Efficacy and Safety in Patients With Moderate Plaque Psoriasis (UNVEIL) (ClinicalTrials.gov: NCT02425826) is the first prospective randomized, placebo (PBO)-controlled trial to demonstrate the clinical efficacy and safety of an oral systemic treatment (apremilast) in systemic- and biologic-naive patients with moderate plaque psoriasis. Apremilast 30mg twice-daily (APR) was clinically effective and well tolerated during the 16-week, double-blind, PBO-controlled phase. The efficacy and safety results of the open-label APR treatment phase up to Week 52 are presented.

Methods: UNVEIL is a Phase IV, multicenter, randomized, PBO-controlled, double-blind study. Men and women at least 18 years of age with chronic plaque psoriasis for six months before screening were included in the study. Subjects had moderate plaque psoriasis at screening and baseline as defined by BSA of 5 to 10 percent and static Physician’s Global Assessment (sPGA) of 3 (moderate) based on a scale ranging from 0 (clear) to 5 (very severe). Included subjects had no prior exposure to systemic or biologic treatments for psoriasis, psoriatic arthritis, or any other condition that could affect the assessment of psoriasis. Potential subjects with an inflammatory or dermatologic condition, including forms of psoriasis other than plaque psoriasis, were excluded. Persons using topical therapy within two weeks or phototherapy within four weeks of randomization were also excluded. Patients were randomized (2:1) to receive APR or PBO during Weeks 0 to 16; patients in the PBO group were switched to APR at Week 16. All patients continued taking APR through Week 52.

The primary efficacy endpoint was the mean percentage change from baseline at Week 16 in PGAxBSA, which represents the product of sPGA and BSA scores. Overall BSA affected by psoriasis is estimated based on the patient’s palm area, which equates to approximately one percent of total BSA. For the 6-point sPGA for plaques in all involved areas, the severity of erythema, scaling, and plaque elevation were each scored; scores were averaged and rounded to the nearest whole number. The secondary efficacy endpoint was the proportions of patients achieving a 75-percent reduction from baseline in PGAxBSA score (PGAxBSA-75) and the sPGA response, defined as a score of 0 (clear) or 1 (almost clear). Quality of life (QOL) was assessed with the Dermatology Life Quality Index (DLQI). Safety was evaluated based on adverse events (AEs), vital signs, clinical laboratory testing, and complete physical examinations. Efficacy and QOL assessments were conducted for the intent-to-treat (ITT) population, which included all randomized patients; safety assessments were conducted in all randomized patients who received one dose of study medication. Mean percentage change from baseline in PGAxBSA and change from baseline in DLQI total score at Week 16 were compared between APR and PBO using a two-sided analysis of covariance model (ANOVA) with treatment and site as factors and baseline value as covariate. PGAxBSA-75 and sPGA responses at Week 16 were evaluated with two-sided Cochran-Mantel-Haenszel tests stratified by site. Efficacy and QOL parameters at Week 52 were evaluated descriptively. Week 16 and Week 52 APR/APR analyses were performed with the ITT population. Week 52 PBO/APR analyses were performed with the modified ITT population (all patients who entered the APR extension phase). The last-observation-carried-forward methodology was used to impute missing values. Safety assessments were summarized using frequencies and percentages.

Results: A total of 221 patients were randomized to study treatment and constitute the ITT population; 185 patients (84%) completed the PBO-controlled phase (Weeks 0–16) and 136/185 patients (74%) completed the APR treatment phase (Weeks 16–52). Demographics and baseline disease characteristics were generally similar between treatment groups. At baseline, mean DLQI total score was comparable between treatment groups, and the mean pruritus Visual Analog Scale (VAS) score was slightly higher in the PBO group. At Week 16, significantly greater improvement in PGAxBSA occurred in patients receiving APR (-48.1%) versus PBO (-10.2%). At Week 52, improvement was maintained in the APR/APR group and emerged in the PBO/APR group after switching to APR. Significantly more patients treated with APR achieved PGAxBSA-75 response at Week 16 (35.4%) versus PBO (12.3%). PGAxBSA-75 response was maintained in the open-label APR treatment phase. Significantly more patients treated with APR achieved an sPGA score of 0 or 1 at Week 16 (30.4%) versus PBO (9.6%). Long-term sPGA response was maintained with APR treatment in the open-label treatment phase. Improvement in DLQI was significantly greater with APR (-4.8) than PBO (-2.4) at Week 16. DLQI improvement was maintained in patients continuing on APR for up to 52 weeks and developed after patients were switched from PBO to APR. Most AEs were mild or moderate. The most common AEs (reported in 5% of patients in either treatment group during the PBO-controlled period) included diarrhea, headache, nausea, upper respiratory tract infection, decreased appetite, and vomiting.

Conclusion: APR was clinically effective in systemic- and biologic-naive patients with moderate plaque psoriasis (BSA of 5–10%). APR significantly improved PGAxBSA score, PGAxBSA-75 response rate, sPGA 0 or 1 response rate, and DLQI total score at Week 16 compared with PBO. Clinical responses were maintained with continued APR treatment through Week 52 and emerged in patients who switched from PBO to APR at Week 16. The incidence of AEs, based on EAIR per 100 patient-years, did not increase with longer exposure to APR. Safety and tolerability were consistent with previous studies; no new safety or tolerability issues were observed up to 52 weeks.

Funding/Disclosures: The authors acknowledge financial support for this study from Celgene Corporation. Bruce Strober receives honoraria as a consultant, payments (to the University of Connecticut) as an investigator, and is an advisory board member of Celgene Corporation. Seth Forman receives research support from the Celgene Corporation. Jerry Bagel is an advisory board member, speaker, consultant, and/or receives research support from the Celgene Corporation. Mark Lebwohl is an investigator and/or consultant for the Celgene Corporation. J. Mark Jackson receives research, honoraria, consulting, and/or other support from Celgene Corporation. Joana Goncalves and Eugenia Levi are employees of the Celgene Corporation. Kristina Callis Duffin is a consultant, steering committee member, advisory board member, has received grants, and/or has received honoraria from the Celgene Corporation.

Improvement in scalp and nails with apremilast in patients with moderate plaque psoriasis naive to systemic and biologic therapy: 52-week results of the UNVEIL study.

Improvement in scalp and nails with apremilast in patients with moderate plaque psoriasis naive to systemic and biologic therapy: 52-week results of the UNVEIL study.

Presenters: Jackson JM1, Alikhan A2, Lebwohl M3, Stein Gold L4, Levi E5, Bagel J6,

Affiliations: 1University of Louisville, Forefront Dermatology, Louisville, KY; 2University of Cincinnati, Department of Dermatology, Cincinnati, OH; 3Icahn School of Medicine at Mount Sinai, New York, NY; 4Henry Ford Health System, West Bloomfield, MI; 5Celgene Corporation, Summit, NJ; 6Psoriasis Treatment Center of Central New Jersey, East Windsor, NJ

Background/Objective: Patients with moderate plaque psoriasis (i.e., 5–10% psoriasis-involved Body Surface Area [BSA]) often receive no treatment or are undertreated with topical monotherapy. Plaque psoriasis that occurs in difficult-to-treat areas such as the scalp and nails might be disproportionately more distressing to patients because it is highly visible and can severely impact daily functioning. Apremilast is an oral, small-molecule phosphodiesterase 4 inhibitor that works intracellularly to regulate inflammatory pathways relevant to the pathogenesis of psoriasis. Apremilast has been shown to be effective and has demonstrated acceptable tolerability in Phase IV clinical studies in patients with moderate-to-severe psoriasis. Evaluating Apremilast in a Phase IV Trial of Efficacy and Safety in Patients With Moderate Plaque Psoriasis (UNVEIL; ClinicalTrials.gov: NCT02425826) is the first prospective, randomized, placebo (PBO)-controlled trial to demonstrate the clinical efficacy and safety of a systemic treatment (apremilast) in systemic- and biologic-naive patients with moderate plaque psoriasis. To further understand the efficacy of apremilast in patients with moderate plaque psoriasis, analyses were performed in the subset of patients with baseline scalp and/or nail involvement.

Methods: UNVEIL is a Phase IV, multicenter, randomized, PBO-controlled, double-blind study. Men and women at least 18 years of age with chronic plaque psoriasis for six months before screening were included in the study. Subjects had moderate plaque psoriasis at screening and baseline as defined by BSA of 5 to 10 percent and static Physician’s Global Assessment (sPGA) of 3 (moderate) based on a scale ranging from 0 (clear) to 5 (very severe). Included subjects had no prior exposure to systemic or biologic treatments for psoriasis, psoriatic arthritis, or any other condition that could affect the assessment of psoriasis. Potential subjects with an inflammatory or dermatologic condition, including forms of psoriasis other than plaque psoriasis, were excluded. Persons using topical therapy within two weeks or phototherapy within four weeks of randomization were also excluded. Patients were randomized (2:1) to receive apremilast 30mg twice daily (APR) or PBO during Weeks 0 to 16; patients in the PBO group were switched to APR at Week 16. All patients continued taking APR through Week 52. A nonmedicated moisturizer was the only topical therapy permitted during the study.

Patients with a baseline Scalp Physician Global Assessment (ScPGA) score of 1 or a Nail Psoriasis Severity Index (NAPSI) score of 1 in the target nail were included in subanalyses of scalp and nail involvement. ScPGA was assessed on a 6-point scale ranging from 0 (clear) to 5 (very severe). One thumbnail or fingernail with the worst nail psoriasis involvement at baseline was designated as the target nail. NAPSI score was calculated in the target nail as the sum of scores for the nail matrix and nail bed, with each score based on the number of quadrants with psoriasis features. Efficacy assessments in patients with scalp psoriasis at baseline included proportion of patients achieving ScPGA score of 0 (clear) or 1 (minimal), with a 2-point reduction from baseline score, at Week 16 and Week 52. Efficacy assessments in patients with nail psoriasis at baseline included change from baseline in NAPSI score at Week 16 and Week 52, proportion of patients achieving a 50-percent reduction from baseline in NAPSI score (NAPSI-50) at Week 16 and Week 52. Safety was evaluated based on adverse events (AEs), vital signs, clinical laboratory testing, and complete physical examinations. Efficacy and safety assessments were conducted for the intent-to-treat (ITT) population, which included all randomized patients; safety assessments included all patients who received one dose of study medication. The proportions of patients achieving ScPGA and NAPSI-50 responses were compared between the PBO and APR groups at Week 16 using a two-sided Cochran-Mantel-Haenszel test stratified by site. Changes from baseline in NAPSI score at Week 16 were compared between treatment groups using a two-way analysis of covariance (ANCOVA) model with treatment and site as factors and baseline value as a covariate. Efficacy parameters at Week 52 were evaluated descriptively. The last-observation-carried-forward (LOCF) methodology was used to impute missing values. Safety assessments were summarized using frequencies and percentages.

Results: A total of 221 patients were randomized to study treatment. Of these, 167 patients (75.6%) had scalp psoriasis and 83 patients (37.6%) had nail psoriasis at baseline. Demographic and baseline disease characteristics of the cohorts with scalp psoriasis or nail psoriasis were generally comparable between treatment groups. Mean baseline NAPSI scores were comparable between treatment groups. Across treatment groups, most patients had mild or moderate scalp involvement (i.e., ScPGA score of 2 or 3). Regarding efficacy of APR in scalp psoriasis, at Week 16, more patients treated with APR than with PBO achieved an ScPGA score of 0 or 1 with a 2-point reduction from baseline (38.4% vs. 20.0%, P=0.0178). At Week 52, patients remaining on APR maintained ScPGA response, and those who switched to APR from PBO at Week 16 (PBO/APR) demonstrated an improvement in ScPGA response comparable to those who continued APR treatment (APR/APR). An ScPGA score of 0 or 1 with a 2-point reduction from baseline was achieved by 47.7 percent of patients randomized to APR who continued on APR (APR/APR), and 46.9 percent of patients randomized to PBO who switched to APR (PBO/APR). Regarding the efficacy of APR in nail psoriasis, at Week 16, the mean percentage change from baseline in NAPSI score was -10.5 percent in the PBO group and -28.9 percent in the APR group (P=0.12). At Week 52, continued improvement in NAPSI score was seen in patients who remained on APR treatment (mean percentage change from baseline, -51.9%). Patients who switched from PBO to APR at Week 16 demonstrated an improvement in NAPSI score (mean percentage change from baseline, -52.7%). At Week 16, NAPSI-50 response was achieved by 18.5 percent of patients in the PBO group and 26.8 percent of patients in the APR group (P=0.50). Although differences in NAPSI-50 response with APR compared with PBO are numerically greater, the number of patients with nail psoriasis at baseline was low and thus statistical significance was not demonstrated. At Week 52, the proportion of patients who achieved NAPSI-50 response increased in patients who remained on APR treatment and in patients who switched to APR from PBO at Week 16.

The most common AEs reported with APR treatment from 0 to 52 weeks were diarrhea, nausea, headache, and nasopharyngitis. Most AEs were mild or moderate; discontinuations due to AEs occurred in 6.6 percent of patients over the 52-week study. The incidence of AEs, based on exposure-adjusted incidence rate (EAIR) per 100 patient-years, did not increase with longer exposure up to 52 weeks when compared with Weeks 0 to 16. No new safety or tolerability issues were observed up to 52 weeks.

Conclusion: APR treatment improved scalp and nail psoriasis at Week 16 in systemic- and biologic-naive patients with moderate plaque psoriasis (BSA 5–10%); continued improvement was seen with APR treatment up to 52 weeks. The efficacy of APR on scalp and nail psoriasis was consistent with that previously reported in patients with moderate-to-severe plaque psoriasis in randomized Phase III trials. The safety and tolerability profile of APR was also consistent with previous studies.

Funding/Disclosures: The authors acknowledge financial support for this study from Celgene Corporation. J. Mark Jackson provides research, honoraria, consulting, and/or other support to the Celgene Corporation. Ali Alikhan is a former speaker for Celgene Corporation. Mark Lebwohl is an investigator and/or consultant for the Celgene Corporation. Linda Stein Gold is an investigator and/or consultant for the Celgene Corporation. Joana Goncalves and Eugenia Levi are employees of the Celgene Corporation. Jerry Bagel is an advisory board member, speaker, consultant, and/or provides research support to the Celgene Corporation.

Sustained improvement in patient-reported outcomes with continued apremilast treatment over 104 weeks in patients with moderate-to-severe psoriasis

Sustained improvement in patient-reported outcomes with continued apremilast treatment over 104 weeks in patients with moderate-to-severe psoriasis

Presenters: Chapman S1, Cirulli J2, McBride S3

Affiliations: 1Dartmouth–Hitchcock Medical Center, Lebanon, NH; 2Celgene Corporation, Summit, NJ; 3Royal Free London NHS Foundation Trust, London, UK

Background/Objective: Psoriasis is a chronic, systemic, inflammatory disease that is associated with significant impairments in quality of life (QoL), which can include physical discomfort, pruritus, and emotional distress. Apremilast is an oral, small-molecule phosphodiesterase 4 inhibitor that has demonstrated efficacy and safety versus placebo (PBO) in the LIBERATE global Phase IIIb trial in patients with moderate-to-severe plaque psoriasis. Efficacy was maintained for up to 104 weeks in patients who continued treatment with apremilast 30mg twice-daily (APR) in the LIBERATE trial. To further understand the clinical profile of APR, the effect of long-term APR treatment on patient-reported outcomes assessed at 104 weeks was evaluated in the LIBERATE patient population.

Methods: Adults 18 years of age or older with chronic plaque psoriasis for at least 12 months and who were candidates for phototherapy with no prior exposure to biologics for the treatment of psoriatic arthritis or psoriasis were included. Included patients had moderate-to-severe plaque psoriasis, as defined by Psoriasis Area and Severity Index (PASI) score 12, psoriasis-involved Body Surface Area (BSA) 10 percent, and static Physician Global Assessment (sPGA) score 3. Patients included in the study had inadequate response, inability to tolerate, or contraindication to one conventional systemic agent for the treatment of psoriasis. Patients who were excluded had prior treatment with more than three systemic agents for the management of psoriasis, other clinically significant or major uncontrolled diseases, and/or serious infections, including latent, active, or history of incompletely treated tuberculosis. LIBERATE consisted of two treatment phases: a 16-week randomized, double-blind, PBO-controlled phase and an 88-week APR extension phase for an overall treatment duration of 104 weeks. Patients were randomized (1:1:1) to PBO, APR, or etanercept 50mg once-weekly (ETN). At Week 16, all patients in the PBO and ETN groups switched to APR, and patients in the APR group continued APR. Treatment with APR was maintained from Weeks 16 to 104 (APR extension phase).

At Weeks 16 and 104, the proportion of patients achieving response, defined as the minimal clinically important difference (MCID), was evaluated for the following patient-reported outcomes: 1) Dermatology Life Quality Index (DLQI) MCID defined as an at least 5-point decrease from baseline in patients with baseline DLQI score over 5; 2) pruritus Visual Analog Scale (VAS; 0–100mm) MCID defined as an at least 20 percent decrease from baseline; 3) 36-Item Short Form Health Survey version 2 (SF-36v2) Mental and Physical Component Summary scores (MCS and PCS)–both MCIDs defined as an increase of at least 2.5 points from baseline; and 4) Patient Health Questionnaire-8 (PHQ-8)–MCID defined as achievement of score 4 (no significant depressive symptoms). Safety was assessed based on adverse events (AEs), vital signs, clinical laboratory assessments, and physical examinations. Achievement of MCID on the DLQI at Week 16 and Week 104 was a prespecified exploratory endpoint, whereas achievement of MCID on the pruritus VAS, the MCS and PCS, and the PHQ-8 were post-hoc analyses. All MCID analyses were performed using the modified intent-to-treat (mITT) population, which included all randomized patients who received one dose of study medication and had an evaluation at baseline and at the specified time point. Endpoints were analyzed using descriptive statistics, including proportions of patients achieving each endpoint by treatment group; associated 95-percent confidence intervals (CIs) were calculated. All data were analyzed as observed, with no imputation for missing values. The safety population consisted of all patients who were randomized and received one dose of study medication. Descriptive statistics were used for summaries of treatment-emergent AEs and other safety assessments.

Results: The mITT population consisted of 250 patients (PBO, n=84; APR, n=83; ETN, n=83). Patient demographics and baseline disease characteristics were generally comparable between treatment groups. The proportions of patients achieving the MCID for the MCS were generally similar among the treatment groups at Week 16. At Week 104, MCS response was maintained in PBO/APR patients and was comparable between APR/APR and ETN/APR patients at Week 104. At Week 16, the proportion of patients achieving PCS MCID was lowest in the PBO group. At Week 104, PCS response was comparable between the APR/APR and ETN/APR groups and remained lower in the PBO/APR group.

At Week 16 and Week 104, proportions of patients achieving the MCID for PHQ-8 (i.e., score 4 [no significant depressive symptoms]) were generally similar among the treatment groups; response was maintained at Week 104 in the APR/APR group and in patients who switched at Week 16 from ETN or PBO to APR. During the PBO-controlled period (Weeks 0 to 16), AEs occurring in five percent of patients in any treatment group were diarrhea, nausea, upper respiratory tract infection, nasopharyngitis, headache, and tension headache. During the APR extension phase (Weeks 16–104), AEs that occurred in five percent of patients in any treatment group included those observed during the PBO-controlled period as well as arthralgia, rebound psoriasis, pain in extremity, bronchitis, psoriasis, and sinusitis. Most AEs were mild or moderate in severity, did not increase with prolonged APR exposure, and did not lead to study discontinuation. No clinically meaningful changes were reported in laboratory parameters. No cases of tuberculosis (new or reactivation) were reported during the trial.

Conclusion: In biologic-naive patients with moderate-to-severe psoriasis, improvements in patient-reported outcomes, including QOL and pruritus, were generally maintained with continued APR treatment up to 104 weeks. AEs were consistent with the known safety profile of APR.

Funding/Dislcosures: The authors acknowledge financial support for this study from Celgene Corporation. Joshua Cirulli is an employee of the Celgene Corporation.

Head-to-Head in Psoriasis: Brodalumab vs Ustekinumab

It has been unusual for companies to risk direct comparisons of new agents with recent vintage competitors, but that is beginning to happen in the crowded market for biologic agents in psoriasis.  It is also important to note that the trend toward a higher bar for efficacy assessment of treatments for this disease is continuing with a 100% reduction in the psoriasis area-and-severity index score (PASI 100) being used as the benchmark for efficacy.

In two phase III studies (AMAGINE-2 and AMAGINE-3), patients with moderate-to-severe psoriasis were randomized to brodalumab, ustekinumab, or placebo.

The primary aims were to evaluate the superiority of brodalumab over placebo at week 12 with the primary endpoint being the percent of patients achieving at least a 75% reduction in the psoriasis area-and-severity index score (PASI 75) and a static physician’s global assessment (sPGA) score of 0 or 1 (clear or almost clear skin), as well as the superiority of brodalumab over ustekinumab at week 12 with respect to PASI 100.

At week 12, PASI 100 response rates were significantly higher with 210 mg of brodalumab than with ustekinumab (AMAGINE-2, 44% vs 22%; AMAGINE-3, 37% vs 19%; P<0.001).

Rates of neutropenia were higher with brodalumab and with ustekinumab than with placebo and mild or moderate candida infections were more frequent with brodalumab than with ustekinumab or placebo (Lebwohl, 2015).


References

Lebwohl M, Strober B, Menter A, et al. Phase 3 studies comparing brodalumab with ustekinumab in psoriasis. N Engl J Med. 2015;373:1318-28.

Maui Derm 2016 Highlights: Psoriasis

B. Strober, MD, PhD
C. Leonardi, MD
J. Gelfand, MD
A. Blauvelt, MD
A. Kavanaugh, MD
L. Stein-Gold, MD

Newer Biologic Therapies Show Promise for Psoriasis Relief:

There has been increased focus on the IL-23/IL-17 in psoriasis and its treatment. IL-23 is upstream (targeted by tildrakizumab, guselkumab, and BI 655066), IL-17 is in the middle of the stream (targeted by ixekizumab and secukinumab), and the IL-17 receptor is downstream (targeted by brodalumab).

Efficacy of new targeted treatment options for psoriasis:

  • Gesulkumab has shown excellent efficacy with PASI 75 scores approaching 80%.
  • Tildrakizumab has demonstrated excellent efficacy in a phase II trial. PASI 75 scores in this study ranged from 75-80% for different tildrakizumab doses.
  • BI 655066 has demonstrated remarkable efficacy after a single treatment with 54% of patients achieving and sustaining PASI 100 for 9 months.
  • Secukinumab has demonstrated excellent efficacy (PASI 75 scores in >75% of patients) and a rapid onset of action (3 weeks for a mean reduction of in PASI scores).
  • Ixekizumab has also demonstrated very strong efficacy (>80% of patients achieving PASI 75) and has a very rapid onset of action. It is also numerically superior to adalimumab for the treatment of PsA.

The number needed to treat (NNT) is number of patients who need a specific treatment to gain one additional good outcome (e.g., achievement of PASI 75). The best NNT is 1, where every patient experiences a benefit with the treatment The NNT for achievement of patient achieving PASI 75 is about 1 for both secukinumab and ixekizumab. They are 1.7 and 1.4, respectively, for PASI 90; and 3.6 and 2.8 for achieving PASI 100. These values are far lower (better) than those for non-biologic synthetic agents or TNF inhibitors. These new therapies mean that clearance is an achievable goal for patients with psoriasis.

There are many well-established comorbidities of psoriasis, including cardiovascular and metabolic disease. There is also increased mortality in patients with psoriasis (a 5-year reduction in lifespan). Patients with psoriasis are 30 times more likely to have a cardiovascular event than to develop melanoma. Your patients with psoriasis should be carefully evaluated for these very common conditions

Emerging comorbidities of psoriasis include chronic obstructive pulmonary disease, asthma, and chronic kidney disease. It is important to understand the entire spectrum of disease in each of your patients with psoriasis and ask questions that can uncover depression that may result from this burden.

A key question is how to identify patients with psoriasis likely to develop psoriatic arthritis (PsA). A good question to ask patients with psoriasis is, “Do you have joint pain or other joint symptoms that you want to do something about?”

Tapering therapy is an attractive approach to patient management in patients with PsA and rheumatoid arthritis (RA), but there is some evidence that stopping medication and restarting after a flare may not regain the efficacy attained prior to the treatment holiday.

It is important to understand that patients’ and physicians’ evaluation of the impact of their disease are very different and patient reported outcomes (PROs) are very important for determining “how patients do”.

Variables that significantly affect patients’ response to disease include gender and the portion of the body surface affected by psoriasis. Women are more adversely affected by psoriasis than men and the head and face are the most important parts of the body for influencing the patients’ perception of their disease severity.

It is important to keep in mind features of therapy that can affect real-world patient adherence, Maintenance regimens that are less complicated enhance the potential for increased patient adherence and successful outcomes.

Psoriasis 2015: More Pearls from Bruce Strober, MD, PhD

  • Hepatitis B serologies should be obtained prior to starting TNF inhibitors. Understanding the meaning of the various tests is important.
  • Continuous therapy with biologics more reliably guarantees persistent response.
  • Palmoplantar psoriasis is usually more difficult to treat, yet most effective psoriasis medications can be successful in at least 1/3 of patients.
  • Subcutaneous methotrexate demonstrates better bioavailability, tolerability, and efficacy when compared to oral methotrexate.
  • Ustekinumab is approved for the signs and symptoms of psoriatic arthritis, yet displays lesser efficacy for this disease than that of the TNF inhibitors.
  • All biologics have demonstrated efficacy for the treatment of nail psoriasis.
  • It is safe to use TNF-inhibitors in the context of hepatitis C infection.
  • Immunogenicity is one reason biologic therapies lose efficacy over time.
  • Biologic therapies have not been demonstrated to cause solid tumor malignancies at a rate greater than the baseline rate of patients with psoriasis.
  • Patients with a prior history of malignancy often can safely receive biologic therapies.
  • Live vaccines should only be administered to patients who are not currently receiving biologic therapies.
  • TNF inhibitors are thought to be safe to use during pregnancy and breastfeeding.

Psoriasis 2015: Pearls from Bruce Strober, MD, PhD

  • Apremilast achieves PASI 75 in approximately 30% of patients after 16 weeks of therapy.
  • Apremilast has FDA-approval for the treatment of both psoriasis and psoriatic arthritis.
  • Apremilast is associated with a >5% weight loss in between 10-20% of treated patients.
  • Secukinumab is the FDA-approved biologic medication with currently the highest level of clinical efficacy of all the self-injectable medications.
  • A side effect of secukinumab is oral candidiasis, which is usually mild and occurs in up to 5% of patients.
  • Secukinimab therapy should be used with caution in patients with inflammatory bowel disease.
  • IL-23 inhibition is a future mechanism of action for psoriasis therapy that shows very high efficacy in early clinical trials.
  • Ixekizumab is an IL-17 inhibitor that promises the highest level of efficacy of its class.
  • Tofacitinib inhibits JAK kinases and likely will be an oral drug approved for psoriasis. The only question is which dose, 5 or 10 mg, will be approved.
  • Secukinumab, ustekinumab and apremilast, like TNF inhibitors, are able to treat psoriatic arthritis to varying levels of efficacy. The TNF inhibitors remain the gold-standard therapies for psoriatic arthritis, though.

Psoriatic Arthritis: Key Developments and Future Directions

Arthur Kavanaugh, MD

Dr Kavanaugh provides us with his clinical pearls on psoriatic arthritis (PsA)…

  • There is increasing evidence that early diagnosis and treatment of PsA results in improved outcomes
  • There exists a large gap and unmet need in PsA, with many patients not being evaluated by doctors or receiving appropriate therapy
  • Because skin manifestations usually precede joint involvement, often by years, Dermatologists play a key role in PsA diagnosis. However, this can present challenges.
  • New guidelines for PsA treatment are under development, and may provide some assistance to clinicians.
  • TNF inhibitors have allowed improved outcomes in PsA, and there continues to be great interest in optimizing therapy with these agents.
  • There is great interest in new targets and agents for the treatment of PsA. Recently revealed data with IL-17 inhibition show promise for treatment of all the various domains of PsA, including peripheral arthritis, skin and nail disease, enthesitis and dactylitis, and axial/spinal arthritis.
  • The IL-12/23 inhibitor ustekinumab was approved last year in PsA and has been shown to be effective across domains of disease.
  • The PDE4 inhibitor apremilast received FDA approval for PsA 3/21/14 and for psoriasis 9/23/14. Its use is increasing in the clinic, for diverse PsA patients. Safety is a particularly attractive feature of this drug.
  • Additional agents are in development for PsA.
  • Optimal management of PsA depends on the levels of activity and severity across the various domains of disease.

Psoriasis 2015: Clinical Pearls

Bruce Strober, MD, PhD

Dr Strober provides us with some important takeaway points from his psoriasis lecture…

  • Apremilast achieves PASI 75 in approximately 30% of patients after 16 weeks of therapy.
  • Apremilast has FDA-approval for the treatment of both psoriasis and psoriatic arthritis.
  • Apremilast also has been shown to provide improvement for nail and scalp psoriasis, and the reduction of pruritus.
  • Apremilast is associated with a >5% weight loss in between 10-20% of treated patients.
  • Secukinumab is the FDA-approved biologic medication with currently the highest level of clinical efficacy of all the self-injectable medications.
  • A side effect of secukinumab is oral candidiasis, which is usually mild and occurs in up to 5% of patients.
  • IL-23 inhibition is a future mechanism of action for psoriasis therapy that shows very high efficacy in early clinical trials.
  • Tofacitinib inhibits JAK kinases and will be an oral drug approved for psoriasis.
  • Tofacitinib increases the risk for varicella-zoster.
  • Both secukinumab, apremilast and tofacitinib are all drugs that variably treat psoriatic arthritis.