What is the approval history and clinical development pathway of Skyrizi?

7 March 2025
Introduction to Skyrizi

Overview of Skyrizi Skyrizii (risankizumab) is a humanized immunoglobulin G1 monoclonal antibody that selectively targets the p19 subunit of interleukin‑23 (IL‑23). By binding to this specific subunit, Skyrizi interrupts the IL‑23-dependent inflammatory cascade while leaving IL‑12 largely unaffected. This mechanism of action is thought to be critical in modulating inflammatory responses in various immune-mediated diseases. The drug’s highly specific targeting profile has demonstrated significant efficacy across multiple indications, lessening the burden of chronic inflammatory conditions. Skyrizi’s formulation allows both intravenous and subcutaneous administration depending on the indication, which offers flexibility in dosing regimens and is an appealing feature from a patient convenience perspective.

Therapeutic Indications
Skyrizi is primarily approved for the treatment of moderate-to-severe plaque psoriasis in adults who are candidates for systemic therapy or phototherapy. In addition to plaque psoriasis, Skyrizi has received regulatory approval in several regions for the treatment of active psoriatic arthritis in adults. More recently, it has emerged as a treatment option in inflammatory bowel diseases, particularly Crohn’s disease. In Japan, for example, Skyrizi is approved not only to manage plaque psoriasis and psoriatic arthritis but also erythrodermic psoriasis in patients who have shown an inadequate response to conventional therapies, as well as for both induction and maintenance in moderately to severely active Crohn’s disease. Its diverse therapeutic indications underscore its role as a versatile immunotherapy, backed by robust clinical data across different patient populations.

Regulatory Approval History

Initial Approval
Skyrizi’s journey began with its initial global approval in 2019. The first regulatory nod was secured in Japan in March 2019 for the treatment of psoriasis vulgaris, psoriatic arthritis, generalized pustular psoriasis, and erythrodermic psoriasis. This early approval stemmed from positive Phase III trial results that established the efficacy of Skyrizi in psoriasis – notably a significant improvement in the Psoriasis Area and Severity Index (PASI 90 and PASI 100 responses) compared to placebo and even some established therapies. Following the first approval in Japan, the drug rapidly gained momentum as regulatory authorities in other major markets reviewed and subsequently approved Skyrizi. In the United States, the Food and Drug Administration (FDA) approved Skyrizi for the treatment of moderate-to-severe plaque psoriasis in 2019, marking a pivotal milestone in its development trajectory. This US approval was critical not only because of the market size but also because it validated the novel mechanism of action of IL‑23 inhibition in a highly competitive therapeutic landscape.

Subsequent Approvals and Indications
Building upon its initial success, regulatory approvals for Skyrizi expanded in the following years. After the foundational approval for plaque psoriasis, subsequent approvals targeted additional indications. For instance, Skyrizi secured approval for the treatment of active psoriatic arthritis in adults. The extension beyond skin manifestations to include joint involvement highlighted the drug’s broad anti‑inflammatory capability and provided a new treatment option for patients who previously had limited choices.

Later, as clinical data matured, Skyrizi entered the inflammatory bowel disease space, particularly for Crohn’s disease. In the United States, Skyrizi was formally approved for Crohn’s disease in June 2022, following robust Phase III data from studies such as ADVANCE, MOTIVATE, and head-to-head comparator trials that established both induction and maintenance efficacy. The European Medicines Agency’s (EMA) recent positive recommendation from CHMP for treating moderately to severely active Crohn’s disease further solidified its emerging role in gastrointestinal indications. With these approvals in place, Skyrizi became one of the rare agents with multiple indications spanning dermatology, rheumatology, and gastroenterology, contributing to a diversified portfolio that reduces reliance on any single therapeutic segment.

Clinical Development Pathway

Phase I Clinical Trials
The clinical development of Skyrizi began with Phase I trials, designed primarily to assess its safety profile, pharmacokinetics, and pharmacodynamics in healthy volunteers and selected patient populations. Early phase studies were instrumental in determining the appropriate dosing regimens as well as the tolerability of risankizumab when administered via both intravenous and subcutaneous routes. These initial trials established that the drug had an acceptable safety profile and predictable pharmacokinetic behavior, which laid the groundwork for subsequent dose‑finding studies in patients with active disease. Moreover, Phase I data were also valuable in confirming the mechanism of action – the selective binding to IL‑23 p19 – which provided the rationale for its later use across a spectrum of inflammatory diseases.

Phase II Clinical Trials
Following the promising outcome of Phase I studies, Skyrizi transitioned into Phase II clinical trials. These studies focused on dose ranging and initial assessments of efficacy in the target patient populations. In psoriasis patients, Phase II trials demonstrated significant improvements in skin clearance as measured by the PASI score. Early signals indicated that risankizumab induced marked improvements compared to placebo, with dose-dependent increases in the proportion of patients achieving higher levels of skin clearance (such as PASI 75, PASI 90, and even PASI 100 responses).

Phase II studies also contributed critical data concerning the drug’s safety with longer exposure and helped refine the dosing schedules. This was particularly important as the drug was being considered for both induction and maintenance therapy in chronic conditions. In addition to psoriasis, early Phase II trials began to explore the efficacy of Skyrizi in other immune‑mediated conditions such as Crohn’s disease. These trials provided preliminary evidence that the IL‑23 blockade translated into improvements in clinical remission rates, endoscopic response, and other endpoints, thereby setting the stage for Phase III evaluations. The tolerability and robust efficacy signals observed in Phase II studies played a key role in designing the larger, pivotal Phase III trials that would be critical for regulatory submissions.

Phase III Clinical Trials
Phase III trials were the cornerstone of Skyrizi’s clinical development program. In the arena of moderate-to-severe plaque psoriasis, multiple pivotal Phase III trials—such as UltIMMa‑1, UltIMMa‑2, IMMvent, and IMMhance—were conducted to compare Skyrizi against placebo, as well as active comparators such as ustekinumab (Stelara) and adalimumab (Humira). These trials consistently demonstrated that a higher proportion of patients achieved PASI 90 and even PASI 100 responses when treated with Skyrizi compared to both placebo and active comparators. The durability of these responses was also highlighted in long‑term extension studies, such as LIMMitless, which confirmed the sustained efficacy and safety profile of risankizumab with continuous treatment over several years.

In parallel with studies in plaque psoriasis, Phase III trials were instrumental in evaluating Skyrizi in Crohn’s disease. For instance, the ADVANCE and MOTIVATE studies provided compelling data that showed significant differences between risankizumab- and placebo-treated patients in achieving both clinical remission and endoscopic response at predefined time points. The Phase III SEQUENCE trial, a head-to-head study against Johnson & Johnson’s Stelara, further underscored Skyrizi’s efficacy over an established treatment option in Crohn’s disease. In this trial, at week 24, Skyrizi achieved clinical remission rates of 59% compared to 40% with Stelara, and at week 48, endoscopic remission rates were 32% for Skyrizi versus 16% for Stelara. These statistically significant differences across co-primary and secondary endpoints, including steroid‑free remissions and endoscopic improvements, provided the key evidence necessary for regulatory bodies in the US and Europe to extend the indication of Skyrizi to Crohn’s disease.

The robust data emerging from Phase III trials, not only in skin-related endpoints but also in gastrointestinal outcomes, confirmed that IL‑23 inhibition is an effective strategy in addressing immune-mediated inflammation across multiple organ systems. Compared to conventional anti‑TNF therapies, Skyrizi exhibited a favorable efficacy and safety profile, which contributed to its positioning as a potential successor for older, established therapies like Humira—especially in the context of declining sales due to biosimilar competition. Overall, the comprehensive Phase III clinical trials provided a solid foundation upon which regulatory agencies recognized the clinical utility of Skyrizi, leading to its approval across multiple indications.

Impact and Future Directions

Current Market Position
Following its multiple regulatory approvals, Skyrizi now occupies a prominent position in the immunology portfolio of AbbVie. Market performance shows rapidly growing sales figures with significant year-over-year revenue increases. For example, recent reports have noted that Skyrizi achieved sales of several billion dollars in the first half of recent years, with continued growth driven by its expanding indication list, including plaque psoriasis, psoriatic arthritis, and Crohn’s disease. Its strong efficacy signals—demonstrated in head-to-head clinical studies against competitors such as Cosentyx, Stelara, and Otezla—have also contributed to increased clinician confidence and wider adoption in clinical practice.

In dermatology, Skyrizi has emerged as a preferred treatment option due to its high rates of skin clearance, favorable dosing schedule of quarterly administration after initial induction, and sustained efficacy over time. Additionally, its safety profile compares favorably with that of older treatments, giving it a competitive advantage in a crowded market. The drug’s effectiveness in achieving higher PASI scores (such as PASI 90 and PASI 100) has often been highlighted in clinical publications and at dermatology congresses, further cementing its status as a next-generation biologic therapy.

Moreover, the impact of Skyrizi extends beyond dermatology. Its recent approval for Crohn’s disease is particularly noteworthy given the challenging therapeutic landscape in inflammatory bowel disease. With key studies demonstrating its superiority in both clinical and endoscopic endpoints compared to established therapies like Stelara, Skyrizi is poised to play a crucial role in the evolving management strategies for Crohn’s disease, which could also translate into broader market acceptance and usage in gastroenterology.

Ongoing Research and Future Prospects
Looking toward the future, ongoing research on Skyrizi continues to explore expanded indications, combination therapy approaches, and long-term outcomes. Several post-marketing studies and open-label extension trials are being conducted to further define the long-term safety, durability of response, and patient-reported outcomes associated with extended Skyrizi therapy. In dermatology, head-to-head comparisons with emerging biologics and small-molecule agents are being planned or are in progress, ensuring that the market position of Skyrizi remains robust as treatment paradigms evolve.

Beyond its established indications, there is ongoing research aimed at identifying novel biomarkers that might predict treatment response or inform personalized dosing strategies. This translational research is particularly important in the context of high variability in treatment responses observed among patients. With the advent of precision medicine, future studies may leverage genomic and proteomic data to optimize treatment regimens for individual patients, thereby enhancing the overall cost-effectiveness and clinical success of Skyrizi.

Furthermore, the success of Skyrizi in treatment-naïve as well as treatment-experienced patients—such as in those who have shown suboptimal responses to IL‑17 inhibitors like Cosentyx and Taltz—opens the possibility for its use in earlier lines of therapy or in combination treatment regimens. This would be significant in patient populations where rapid and sustained disease control is paramount.

In the competitive landscape of biologics, ongoing clinical trials are also exploring the potential of Skyrizi in other immune‑mediated inflammatory diseases. The expanding spectrum of diseases targeted by IL‑23 inhibitors suggests that future indications could include additional subsets of inflammatory arthritis, other gastrointestinal conditions, and possibly even novel dermatological indications. As biosimilars of older drugs like Humira begin to erode market share, successful drugs like Skyrizi that offer differentiated efficacy and safety profiles will likely benefit from favorable prescribing trends and market penetration.

Research into optimizing dosing intervals and exploring alternative routes of administration continues as well. Such studies aim to enhance patient adherence and improve the convenience of treatment, which are important factors in chronic diseases with lifelong treatment requirements. Ongoing safety surveillance and real-world evidence studies will provide additional data on the long-term risk–benefit profile, further informing clinical practice and potentially spurring future regulatory updates regarding dosing and administration guidelines.

Conclusion
In summary, Skyrizi’s development and regulatory approval history represent a comprehensive and multi-staged process that underscores the importance of rigorous clinical evaluation across multiple phases. Initially approved in 2019—first in Japan and subsequently in major markets like the United States—Skyrizi gained rapid acceptance for its efficacy in treating moderate-to-severe plaque psoriasis. Its mechanism of action, targeting the IL‑23 p19 subunit, not only provided impressive clinical outcomes in skin-related endpoints but soon translated into efficacy in other immune‑mediated conditions such as psoriatic arthritis and Crohn’s disease.

The clinical development pathway of Skyrizi began with crucial Phase I studies that established the drug’s safety and pharmacokinetics. Phase II trials then refined dosage and revealed promising efficacy signals, which paved the way for a series of robust Phase III trials in psoriasis and inflammatory bowel disease. These pivotal trials compared the drug against placebo as well as active comparators, demonstrating significant improvements in PASI scores, clinical remission rates, and endoscopic responses. Notably, head-to-head studies such as the SEQUENCE trial against Stelara provided compelling evidence of Skyrizi’s superiority in the treatment of Crohn’s disease.

Market impact following regulatory approvals has been substantial, with Skyrizi establishing itself as a key asset in AbbVie’s immunology portfolio. Its strong performance in both dermatological and gastrointestinal indications, coupled with positive safety profiles and the convenience of a quarterly dosing regimen, has positioned it favorably against competitors. Looking forward, ongoing research is focused on expanding its indications, optimizing treatment strategies, and leveraging biomarkers for personalized medicine. There is optimism that further studies will not only consolidate but also expand its therapeutic footprint across additional inflammatory conditions.

In conclusion, Skyrizi’s approval history and clinical development pathway—from initial Phase I safety studies, through comprehensive Phase II dose-ranging trials, to pivotal Phase III trials leading to multiple approvals—demonstrate a well-orchestrated, evidence-based approach that has positioned the drug at the forefront of next-generation therapies for immune‑mediated diseases. The continued evolution of its research pipeline and market adoption suggests that Skyrizi will remain a leading therapeutic option with significant future potential in both dermatology and gastroenterology, reaffirming its role in transforming treatment paradigms for chronic inflammatory conditions.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成