Interleukin 23 Inhibitors


Questions

  • Q29.1 What are the two subunits that make up interleukin (IL)-23? (Pg. 321)

  • Q29.2 What subunit of IL-23 do IL-23 inhibitors target? (Pg. 321)

  • Q29.3 What interleukin inhibitors are directed against IL-23? (Pg. 321)

  • Q29.4 How does the efficacy of IL-23 inhibition alone compare with IL-12/23 inhibition? (Pg. 321)

  • Q29.5 What conclusions were drawn from the phase III guselkumab trials with regard to efficacy? (Pg. 322)

  • Q29.6 What specific safety concerns (if any) have been elucidated for the IL-23 inhibitors? (Pgs. 324, 326, 327)

  • Q29.7 What conclusions were drawn from the phase III tildrakizumab trials with regard to efficacy? (Pg. 325)

  • Q29.8 What conclusions were drawn from the phase III risankizumab trials with regard to efficacy? (Pg. 326)

  • Q29.9 In what other diseases may IL-23 inhibitors may have utility? ( Table 29.4 ) (Pg. 328)

  • Q29.10 What drugs are ‘in the pipeline’ (no phase III clinical data) that target the IL-23 pathway? ( Table 29.4 ) (Pg. 328)

Abbreviations used in this chapter

Ab

Antibody

ADA

Antidrug antibodies

ADM

Adalimumab

AE

Adverse effects/events

CD

Crohn disease

DLQI

Dermatology Life Quality Index

ETA

Etanercept

FDA

Food and Drug Administration

Grp

Group

GUS

Guselkumab

IGA

Investigator’s Global Assessment

IL

Interleukin

IV

Intravenous

NR

Not reported

PASI

Psoriasis Area and Severity Index

PBO

Placebo

PGA

Physician’s Global Assessment

PS

Psoriasis

PSSA

Psoriasis Symptoms and Signs Diary

RIS

Risankizumab

SAE

Severe adverse effects/events

SC

Subcutaneous

TIL

Tildrakizumab

UC

Ulcerative colitis

UST

Ustekinumab

Introduction

Q29.1 Interleukin (IL)-23 is a heterodimeric cytokine that shares a common p40 subunit with IL-12, yet has a distinct p19 subunit. Since its discovery, IL-23 has emerged as an important inflammatory cytokine in the pathogenesis of psoriasis, particularly in the T helper (Th)17 pathway.

Q29.2 Q29.3 Q29.4 The development of biologic agents directed against both IL-23 and IL-12 have demonstrated promising effects for psoriasis and other autoimmune diseases. However, Lee and colleagues demonstrated elevated levels of the p40 subunit (shared by IL-12 and IL-23) and the p19 subunit (IL-23 specific) but normal levels of the p35 subunit (IL-12 specific) in psoriatic skin lesions. Moreover, Kopp and coworkers reported clinical proof that specific targeting of IL-23p19 improves symptoms of moderate-to-severe plaque psoriasis in a phase I study of tildrakizumab (Ilumya/MK-222 SunPharma). Further, the results from clinical trials comparing IL-23 inhibitors guselkumab (Tremfya/CNTO-1959 Janssen) and risankisumab (BI-655066 Abbvie) with IL-12/23 inhibitor ustekinumab suggest that biologics directed specifically against IL-23 may be more effective than ustekinumab. Therefore, although they share a common subunit, IL-23 may play a more prominent role than IL-12 in the pathogenesis of psoriasis.

Biologics targeting IL-23 have been one of the latest developments in psoriasis treatment. Three biologic agents that specifically bind the p19 subunit of IL-23 have been developed for use in psoriasis and studied in phase III clinical trials: guselkumab, tildrakizumab, and risankizumab ( Tables 29.1 and 29.2 ).

Table 29.1
IL-23 Inhibitors
Generic Name Trade Name Generic Available Manufacturer How Supplied Standard Dosage Range
Guselkumab Tremfya No Janssen 100 mg/mL—prefilled syringe 100 mg at week 0, 4, then every 8 weeks
Tildrakizumab Ilumya No SunPharma 100 mg/mL—prefilled syringe 100 mg at weeks 0, 4, then every 12 weeks
Risankizumab Skyrizi No AbbVie 75 mg/mL—prefilled syringe (x2/150 mg dose) 150 mg at weeks 0, 4, then every 12 weeks

Table 29.2
Key Pharmacology Concepts
Absorption and bioavailability Elimination
Drug Name Peak Levels (Days) Bioavailablity (%) Protein Binding Half-Life (Days) Metabolism Excretion
Guselkumab 5.5 49 NR 15–18 Not elucidated, believed to be proteolysis NR
Tildrakizumab 6 50 mg—80
200 mg—73
NR 23 Not elucidated, believed to be proteolysis NR
Risankizumab 4–10 89 NR 20–28 By proteolysis similar to endogenous IgG NR
NR, Not reported.

US Food and Drug Administration Approval

Guselkumab was approved in the United States in July 2017 for the treatment of moderate-to-severe plaque psoriasis. Dosing of guselkumab is 100 mg administered subcutaneously (SC) at week 0, 4, and then every 8 weeks thereafter. The efficacy and safety of guselkumab have been evaluated in three phase III clinical trials thus far (VOYAGE 1, VOYAGE 2, and NAVIGATE) ( Table 29.3 ). As of February 2019, it is currently undergoing trials evaluating use for psoriatic arthritis (NCT03158285, NCT02319759), and trials for use in Crohn disease are forthcoming (NCT03466411).

Table 29.3
Guselkumab Phase III Clinical Trials for Psoriasis
Study Title Total # Pts Dosages Studied Primary Results Conclusions
VOYAGE 1 837
  • 1.

    100 mg SC weeks 0 & 4, then q8w

  • 2.

    PBO SC weeks 0, 4, & 12, with crossover to GUS at week 16

  • 3.

    ADM 80 mg SC week 0, 40 mg at week 1, then 40 mg q2w

91.2% of patient vs. 5.7% in PBO group and 73.1% in ADM group reached PASI-90 at week 16

IGA 0/1 at week 16 in 85.1% of patients on GUS vs. 6.9% & 65.9% on PBO & ADM, respectively

GUS demonstrated superior efficacy vs. ADM and well-tolerated in patients through 1 year
VOYAGE 2 992
  • 1.

    100 mg SC weeks 0 & 4, then q8w

  • 2.

    PBO SC weeks 0, 4, and 12, crossover to medication week 16

  • 3.

    ADM 80 mg SC week 0, 40 mg week 1, then 40 mg q2w

  • 4.

    Responders (patients ≥PASI-90 from baseline) rerandomized to GUS or PBO with GUS after loss of response

  • 5.

    Nonresponders (i.e., patients <PASI-90 from baseline) at week 28 received GUS

70.0% of patients vs. 2.4% and 46.8% in PBO & ADM groups, respectively achieved PASI-90 at week 16.

IGA 0/1 at week 16 in 84.1% of patients on GUS vs. 8.5% in PBO group & 67.7% in ADM group

In ADM nonresponders, PASI-90 & 100 response rates ↑, 66.1% & 28.6%, respectively, at week 48

GUS appears to be an effective, well-tolerated maintenance therapy in patients with psoriasis including ADM nonresponders
NAVIGATE 837
  • 1.

    45/90 mg SC UST week 0 & 4

  • 2.

    At wk 16, nonresponders (IGA ≥2) randomized to GUS 100 mg SC weeks 16, 20, then q8w or UST 45/90 mg SC q12w to week 40 and observed through week 52

Patients on GUS achieved IGA 0/1 at an average of 1.5 visits vs. 0.7 visits in the UST group at week 40 Patients treated with UST who did not achieve IGA of 0/1 by week 16 derived significant benefit from switching to GUS
ADM, Adalimumab; GUS, guselkumab; IGA, investigator’s global assessment; PASI, Psoriasis area and severity index; PBO, placebo; SC, subcutaneous; UST, ustekinumab.

Tildrakizumab was approved by the FDA for use in the United States in March 2018 for moderate-to-severe plaque psoriasis. Recommended dosing is 100 mg by SC injection at week 0, week 4, and then every 12 weeks thereafter. Results of two phase III clinical trials have been published (reSURFACE 1 and 2). Phase II studies are underway for treatment of psoriatic arthritis and ankylosing spondylitis as of February 2019 (NCT03466411, NCT0298070).

Risankizumab was approved in the United States in April 2019 for the treatment of moderate to severe plaque psoriasis. Recommended dosing will be 150 mg at week 0, week 4, and every 12 weeks thereafter. Clinical trial details are in the text/tables pgs. 326 to 328.

Guselkumab

Pharmacology

Pharmacokinetics

Two phase I studies in psoriasis patients have been conducted using ascending single intravenous (IV) and SC dosing to evaluate pharmacokinetics, immunogenicity, safety, and tolerability of guselkumab, both of which showed linear pharmacokinetics. Mean half-life ranged from 12 to 19 days in healthy subjects and 15 to 17 days in patients with psoriasis. The median time to achieve maximum serum concentration after a single SC dose of 100 mg was 3.2 days, with a range of 2.0 to 7.0 days.

Combined analysis of phase II and III studies found a half-life of 18.1 days with steady-state concentrations reached within 12 weeks. In this analysis, contributing factors to pharmacokinetics were assessed, including body weight, age, gender, race, disease severity and duration, comorbidities, medications, smoking status, alcohol use, and positive immune response to guselkumab. Factors affecting apparent volume of distribution included body weight, and factors affecting apparent clearance included body weight, diabetes, and race (nonwhite vs white). The most significant effect was due to body weight, however given efficacy across different body weight subgroups with 100 mg every-8-week dosing, dose adjustment based on body weight is not warranted. Moreover, variations due to diabetes and race were also deemed to be clinically insignificant.

Clinical Use

Efficacy

Q29.5 In a randomized, double-blind, placebo-controlled phase I trial, 10 out of 14 patients who received one dose of 10 mg, 30 mg, or 100 mg achieved Psoriasis Area Severity Index (PASI)-75 through 24 weeks. Moreover, improvements were observed in epidermal thickness and numbers of CD3 + or CD11c + cells at week 1, with statistically significant reductions in epidermal thickness and T-cell counts observed at week 12 for all guselkumab dose groups compared with baseline. Guselkumab was also shown to attenuate Th17 molecular biomarkers in psoriatic skin. Specifically, serum IL-17A levels in patients responding to guselkumab were found to be reduced. A phase II study further evaluated the role of guselkumab in patients with moderate-to-severe plaque psoriasis and found significantly higher proportions of patients on guselkumab achieved Physician Global Assessment (PGA) scores of 0 or 1 as well as greater improvements in quality of life scores compared to placebo and adalimumab, respectively, at week 16.

The long-term efficacy and safety of guselkumab have been evaluated thus far in three major phase III studies in which treatment was compared with placebo and adalimumab, respectively. VOYAGE 1 consisted of an active-comparator period during which guselkumab 100 mg was compared with adalimumab (week 0–48) and a placebo-controlled period (week 0–16), respectively, after which patients taking placebo crossed over to receive guselkumab through week 48. VOYAGE 2 comprised a placebo-controlled period (week 0–16), an active comparator-controlled period (week 0–28), and a randomized withdrawal and retreatment period (week 28–72). In both studies, significantly more patients in the guselkumab treatment groups achieved the primary endpoints of Investigator’s Global Assessment (IGA) 0/1 and PASI-90 than those receiving placebo or adalimumab at week 16. NAVIGATE involved an open-label period during which patients were treated with ustekinumab (week 0–16) followed by a randomized active-treatment period during which patients who did not achieve IGA 0/1 were randomized to either initiate guselkumab or remain on ustekinumab until week 44. The patients then underwent a follow-up period (week 44–60). In this study, the primary endpoint was the number of visits at which patients achieved IGA 0/1 with at least two grade improvement from week 16 from weeks 28 to 40.

In VOYAGE 1, 91.2% of patients receiving guselkumab achieved PASI-90 compared with 73.1% in the adalimumab group and 5.7% in the placebo group at the end of the placebo-controlled period at week 16. Moreover, the percentage of patients achieving IGA 0/1 at week 16 was significantly higher in the guselkumab group compared with placebo and adalimumab, respectively. Efficacy was noted as early as week 2 compared with placebo. Significantly higher rates of responses were maintained at weeks 24 and 48 when compared with adalimumab, as measured by IGA 0/1 and PASI-90 response. At weeks 24 and 48, 50.5% of patients on guselkumab reached complete clearance (IGA 0). Additionally, at the end of the active-comparator period, the greatest responses were seen in patients who were originally randomized to placebo and then randomized to guselkumab, with approximately 80% of patients maintaining PASI-90. Guselkumab was also effective in treating difficult-to-treat areas, including the scalp, hands, and feet. Patient-reported outcomes based on Psoriasis Symptoms and Signs Diary (PSSD) and Dermatology Life Quality Index (DLQI) questionnaires demonstrated guselkumab was superior to placebo at week 16 and adalimumab at weeks 24 and 48, respectively.

In VOYAGE 2, 84.1% of patients receiving guselkumab achieved IGA 0/1 compared with 8.1% of patients receiving placebo ( P < .001) at the end of the placebo-controlled period. In terms of PASI-90 scores, guselkumab demonstrated superior efficacy as early as week 2 with 70.0% of patients in the guselkumab group achieving PASI-90 by week 16 ( P < .001 when compared with placebo). Moreover, guselkumab was superior to adalimumab by proportion of patients achieving PASI-90 and IGA 0/1 at week 16. Responders rerandomized to the maintenance group maintained better responses than those rerandomized to the withdrawal group at the end of the study (week 48). The median time to loss of PASI-90 response was 15.2 weeks (or 23 weeks after last guselkumab dose) in the withdrawal group. In patients who were nonresponders to adalimumab at week 28 and thus initiated guselkumab, the proportion of patients achieving PASI-90 reached 66.1% after 20 weeks of treatment.

In NAVIGATE, patients receiving guselkumab during the active-treatment period achieved IGA scores of 0/1 or at a significantly higher number of visits compared with the ustekinumab group (1.5 vs. 0.7; P < .001). Moreover, patients in the guselkumab group also reached PASI-90 at significantly more visits, and the mean decrease in PASI score overall was also significantly greater than in the ustekinumab group. Response rates in patients receiving guselkumab increased after the first dose, with maximum response between 32 and 36 weeks. Differences between the guselkumab and ustekinumab groups occurred as early as week 2, or 4 weeks postrandomization. At the end of the observation period, a greater percentage of patients on guselkumab achieved both PASI-90 and PASI-100 compared with the randomized ustekinumab group.

In summary, the VOYAGE 1 and 2 and NAVIGATE trials all demonstrate the efficacy of guselkumab in patients with moderate-to-severe psoriasis, especially for patients who have failed to respond to other biologics, such as adalimumab and ustekinumab. Clinical responses were evident as early as week 2 and maintained over 1 year. It has been suggested that guselkumab may have a slower mechanism of action than the IL-17 inhibitors. However, a potential benefit of guselkumab is its dosing regimen of every 8 weeks, which may be more appealing to patients, and thus, may potentially enhance adherence. However, long-term efficacy remains to be seen.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here