What is the mechanism of action of Risankizumab-RZAA?

7 March 2025

Introduction to Risankizumab-RZAA

Risankizumab-RZAA is a humanized immunoglobulin G1 (IgG1) monoclonal antibody that has been specifically developed to target a key cytokine involved in the inflammatory cascade underlying several immune-mediated diseases. Engineered using recombinant DNA technology in Chinese hamster ovary (CHO) cells, Risankizumab-RZAA has a molecular weight of approximately 149 kDa and is designed as a highly specific antagonist of interleukin-23 (IL-23). Unlike broad-spectrum immunosuppressive agents, this therapeutic agent is focused on selectively inhibiting a distinct subunit of IL-23, thus offering a targeted approach in modulating dysregulated immune responses in diseases such as moderate-to-severe plaque psoriasis, psoriatic arthritis, Crohn’s disease, and potentially other inflammatory conditions. The specificity of its design aims not only to maximize clinical efficacy by intercepting the inappropriate activation of inflammatory pathways but also to minimize off-target effects and adverse events, setting it apart from traditional systemic immunosuppressants.

Therapeutic Indications

Originally, Risankizumab-RZAA received regulatory approval for the treatment of moderate-to-severe plaque psoriasis in adults who are candidates for systemic therapy or phototherapy. Over time, the expanding understanding of IL-23’s pivotal role in immune-mediated processes has led to further investigations into its efficacy for other indications such as psoriatic arthritis, Crohn’s disease, and even ulcerative colitis. Clinical trials have also explored its potential benefits in diseases where IL-23 driven Th17 cell activation contributes significantly to the pathogenesis. For instance, the robust effects observed in plaque psoriasis—demonstrated by high rates of total or near‐complete skin clearance in long‐term studies—highlight its capability to significantly reduce the inflammatory markers and symptomatic manifestations of chronic immune-mediated conditions. In essence, Risankizumab-RZAA serves as a promising therapeutic option in a spectrum of diseases characterized by overactivity of the IL-23/Th17 pathway, providing clinicians with an alternative to traditional agents while offering a more refined profile in terms of efficacy and safety.

Mechanism of Action

Molecular Targets

At the core of Risankizumab-RZAA’s mechanism of action is its highly selective binding to the p19 subunit of interleukin-23 (IL-23). IL-23 is a heterodimeric cytokine comprising a unique p19 subunit and a shared p40 subunit (the latter is also present in interleukin-12). Risankizumab-RZAA distinguishes itself by targeting only the p19 subunit, thereby blocking the activity of IL-23 without interfering with IL-12-mediated pathways. This selective inhibition is crucial because IL-23 is instrumental in the differentiation, survival, and activation of Th17 cells—a subset of T helper cells that secrete proinflammatory cytokines such as IL-17A, IL-17F, and IL-22. By intercepting the signal delivered through the IL-23 receptor, Risankizumab-RZAA effectively blocks the downstream cascade that leads to the amplification of inflammatory responses, particularly within the skin and gastrointestinal tract.

In molecular terms, the binding of Risankizumab-RZAA to IL-23p19 prevents the cytokine from interacting with its receptor on immune cells. The IL-23 receptor is predominantly located on the surface of memory T cells and innate lymphoid cells, which, upon activation, normally initiate and perpetuate inflammatory processes. This blockade interrupts the IL-23/Th17 axis, directly impacting the transcription of inflammatory mediators and thereby reducing pathogenic inflammation. Furthermore, because the antibody is engineered as an IgG1, it possesses the inherent potential to engage with immune cells through its Fc region; however, its primary therapeutic action in this context is mediated by the neutralization of IL-23 rather than by direct cytotoxic effects.

Cellular Pathways

The inhibition of IL-23 by Risankizumab-RZAA results in significant downstream effects on several cellular pathways. The IL-23/Th17 pathway is pivotal for the maintenance and expansion of Th17 cells. Under normal circumstances, IL-23 binds to its receptor on T cells, activating intracellular signaling cascades that include STAT3 phosphorylation and other transcription factors crucial for the expression of pro-inflammatory cytokines. These cytokines, particularly IL-17A, IL-17F, and IL-22, are key drivers in the pathogenesis of psoriasis, psoriatic arthritis, and inflammatory bowel diseases. By preventing IL-23 from engaging with its receptor, Risankizumab-RZAA disrupts these signaling cascades, ultimately decreasing the proliferation and activation of Th17 cells.

This process reduces the secretion of downstream inflammatory cytokines, which in turn leads to a decrease in the recruitment and activation of neutrophils and other immune cells that contribute to the inflammatory milieu at disease sites. Additionally, the blockade impacts cellular differentiation processes by influencing the balance between regulatory T cells (Tregs) and effector T cells, promoting a more controlled immune response. The suppression of IL-23-dependent cellular activation and the subsequent reduction in pro-inflammatory cytokine release help to normalize the aberrant immune activity seen in chronic inflammatory states.

Furthermore, by interfering with this pathway, Risankizumab-RZAA not only diminishes the inflammatory load but also contributes to the restoration of tissue homeostasis. The reduced production of proinflammatory cytokines correlates with a decrease in the stimulation of keratinocytes in the skin, leading to an improvement in the hyperproliferative and inflammatory state characteristic of plaque psoriasis. Analogously, in the gastrointestinal tract of patients with Crohn’s disease, the suppression of the IL-23/Th17 axis allows for diminished mucosal inflammation, which is associated with clinical improvement in disease activity. Thus, the modulation of these cellular pathways forms the cornerstone of Risankizumab-RZAA’s therapeutic benefits.

Clinical Implications

Efficacy in Disease Treatment

The precise targeting of the IL-23p19 subunit by Risankizumab-RZAA has significant clinical implications regarding its efficacy in treating immune-mediated diseases. In clinical trials, Risankizumab-RZAA has demonstrated substantial efficacy in patients with moderate-to-severe plaque psoriasis by achieving high percentages of skin clearance as measured by the Psoriasis Area and Severity Index (PASI) and static Physician’s Global Assessment (sPGA). The interruption of the IL-23/Th17 axis correlates with reduced inflammatory signaling, leading to a marked improvement in clinical symptoms and histopathological findings in psoriatic lesions. Moreover, the sustained blockade of this pathway also contributes to a durable response, with many patients maintaining significant improvements over extended treatment periods.

In addition to psoriasis, the mechanism has been investigated in the context of psoriatic arthritis and inflammatory bowel diseases such as Crohn’s disease and ulcerative colitis. The same molecular rationale applies—by curbing the activity of Th17 cells and their inflammatory mediators, Risankizumab-RZAA offers potential benefits in these conditions as well. For example, clinical trials in psoriatic arthritis have reported improvements in joint symptoms alongside skin benefits, reflecting the broad impact of IL-23 pathway modulation on systemic inflammation. This novel approach distinguishes Risankizumab-RZAA from traditional anti-inflammatory or immunosuppressive therapies, which often act in a broader, less selective manner, and are associated with more generalized immunosuppression and a higher incidence of adverse effects.

The efficacy outcomes observed in the clinical development program have been consistently notable, which supports the concept that highly targeted biologic therapy can achieve both remarkable clinical improvements and a favorable benefit–risk profile. Such outcomes have influenced clinical guidelines and treatment algorithms, positioning IL-23 blockade as a first-line consideration for patients with moderate-to-severe psoriasis and related disorders in whom previous therapies have failed or were poorly tolerated.

Comparison with Other Therapies

When compared with other immunomodulatory therapies, Risankizumab-RZAA’s mechanism of action provides several distinct advantages. Traditional systemic therapies such as methotrexate or cyclosporine exert broad-spectrum immunosuppression, which may inadvertently expose patients to a higher risk of infections and other systemic adverse effects. In contrast, Risankizumab-RZAA, with its targeted inhibition of the IL-23p19 subunit, affords a more selective suppression of pathogenic immune responses while largely sparing other components of the immune system. This specificity not only enhances clinical outcomes but also contributes to a reduced safety burden.

Additionally, other biologics that target the IL-12/IL-23 pathway, such as ustekinumab, inhibit both IL-12 and IL-23 by binding to the shared p40 subunit. Although this leads to effective treatment in many cases, the dual blockade may affect additional pathways that are critical for normal immune function. Risankizumab-RZAA’s selective targeting of IL-23p19 spares IL-12-mediated responses, which could be important for immune surveillance and defense against intracellular pathogens. This distinction is particularly relevant in long-term therapy, where maintaining a balance between effective disease control and preserved immune function is paramount.

Furthermore, when compared with tumor necrosis factor (TNF) inhibitors, which have been the cornerstone of therapy for several immune-mediated diseases, the IL-23/Th17 axis blockade offers a complementary mode of action. TNF inhibitors function by blocking a broad pro-inflammatory cytokine, but they might not be as effective in all patients, and some may lose response over time. In contrast, targeting IL-23 provides a different approach that may overcome resistance observed with TNF inhibitors, particularly in patients who have failed multiple lines of therapy. In summary, the mechanistic specificity, sustained efficacy, and favorable safety considerations make Risankizumab-RZAA an attractive therapeutic option relative to other available treatments.

Safety and Regulatory Considerations

Side Effects and Safety Profile

The side effects associated with Risankizumab-RZAA are generally consistent with those observed in studies investigating monoclonal antibodies. Due to its targeted mechanism of action, the incidence of systemic adverse events tends to be lower compared to less selective immunosuppressive agents. In the clinical trials for plaque psoriasis and psoriatic arthritis, the most common adverse reactions reported included injection site reactions, headache, upper respiratory infections, and, less frequently, incidents of arthralgia and fatigue. Importantly, while immunogenicity is a general concern with therapeutic proteins, the design of Risankizumab-RZAA has mitigated significant neutralizing antibody development in many cases, which contributes to its favorable safety profile.

The immunogenicity profile is also carefully monitored during long-term treatment. In patients with psoriasis, for instance, a proportion of subjects developed anti-drug antibodies; however, these events were typically not correlated with significantly diminished clinical responses or increased adverse event profiles. Safety studies, extended over multiple years and involving thousands of patient-years of exposure, have demonstrated that the rates of serious adverse events remain comparably low. As with any monoclonal antibody therapy, there is a potential risk for opportunistic infections, but the selective inhibition of IL-23 appears to minimize these risks relative to broader immunosuppressive therapies.

Regulatory bodies such as the U.S. Food and Drug Administration (FDA) and equivalent agencies in other countries have thoroughly evaluated the safety and efficacy data from Risankizumab-RZAA’s clinical development program. The safety profile—characterized by stable exposure-adjusted adverse event rates over a long duration—has contributed to its approval for use in patients with specific indications. Additionally, pharmacovigilance programs continue to monitor adverse events post-approval to ensure that any emerging safety issues are promptly addressed.

Approval and Regulatory Status

Risankizumab-RZAA was granted accelerated approval for the treatment of moderate-to-severe plaque psoriasis in adults, with its initial approval dating back to March 2019. Since its first approval in Japan and subsequent worldwide regulatory acceptance, the product’s label has been expanded to include additional indications such as psoriatic arthritis and Crohn’s disease, based on robust clinical trial data demonstrating its efficacy and long-term safety. The detailed descriptions observed in the product’s package inserts and summary of product characteristics (SmPC) reflect comprehensive regulatory reviews that affirm both the quality and the safety of the therapeutic agent.

Regulatory review processes have highlighted the benefit–risk balance of Risankizumab-RZAA, emphasizing its targeted mechanism, favorable safety outcomes, and patient tolerability. The inclusion of detailed risk management plans, particularly concerning the monitoring of potential immunogenicity and infection risks, underscores the regulatory commitment to patient safety. Moreover, the design of the clinical trials, which spanned multiple patient populations and extended periods, provided regulators with ample evidence of sustained efficacy and manageable adverse events, ultimately supporting full approval and ongoing use in clinical practice.

Future Research Directions

Ongoing Clinical Trials

The clinical development of Risankizumab-RZAA continues to evolve, with numerous ongoing clinical trials designed to further define its role in various immune-mediated diseases and to optimize dosing regimens. Many of these trials are investigating its long-term efficacy and safety in patients with plaque psoriasis and psoriatic arthritis, but a significant number are also exploring its application in inflammatory bowel diseases (IBDs) such as Crohn’s disease and ulcerative colitis. These studies are assessing not only the clinical response in terms of symptom improvement and quality of life but also leveraging biomarker-driven endpoints such as endoscopic response and histologic improvement.

The design of these studies is often comprehensive, incorporating multiple dosing schedules and comparing Risankizumab-RZAA to both placebo and other established biologics. For example, trials have evaluated endpoints such as the percentage of patients achieving PASI90, PASI100, and sPGA0/1 responses over several weeks to months, providing a detailed assessment of both short-term and long-term efficacy. Additionally, ongoing studies include robust pharmacokinetic and pharmacodynamic evaluations that seek to understand how individual patient factors—such as body weight, baseline disease characteristics, and the presence of anti-drug antibodies—influence response. Such investigations will help personalize therapy and optimize the risk-benefit profile for diverse patient populations.

Beyond efficacy in established indications, some ongoing studies are considering the potential of Risankizumab-RZAA in other autoimmune and inflammatory conditions. This includes exploring its use as part of combination therapy regimens and further delineating its mechanism in modulating the IL-23/Th17 axis. These trials are expected to not only reinforce the observed clinical benefits but also to uncover nuanced insights into which patient subgroups may derive the greatest therapeutic advantage.

Potential for New Indications

While the primary focus of Risankizumab-RZAA has been on dermatologic and gastrointestinal disorders, the precise immune modulation achieved through targeted IL-23 inhibition opens avenues for potential new indications. Recent preclinical and early-phase clinical studies have suggested that other diseases characterized by IL-23/Th17 dysregulation, such as certain forms of rheumatoid arthritis, multiple sclerosis, and perhaps some atypical inflammatory conditions like postpartum depression, could benefit from similar therapeutic approaches.

Given that IL-23 plays a role in the maintenance of inflammatory responses in a range of tissues, further exploration is warranted to determine if modulating this cytokine might provide clinical benefits in diseases where excess inflammation is a driving factor. The evolving understanding of cytokine networks and cellular signaling in autoimmune pathogenesis may reveal additional niches where Risankizumab-RZAA’s targeted mechanism could be exploited. Furthermore, potential combination studies with other monoclonal antibodies or small-molecule inhibitors are under consideration, aiming to enhance pathogenetic suppression by acting on complementary pathways. This integrative approach may yield synergistic effects, particularly in patients who are refractory to monotherapy with existing agents.

Research into these areas is likely to focus on establishing clear clinical endpoints, determining optimal dosing strategies, and ensuring that any potential increase in efficacy does not compromise the already favorable safety profile. With many of these early investigations still in preliminary phases, long-term data will be critical to determining whether the potential new indications can be translated into meaningful, approved therapies.

Conclusion

In summary, Risankizumab-RZAA functions by a highly targeted mechanism focused on neutralizing IL-23 through selective binding to its p19 subunit. This precise interference prevents the activation of key inflammatory cellular pathways driven by Th17 cells, which are responsible for producing potent proinflammatory mediators. By blocking the IL-23/Th17 axis, Risankizumab-RZAA effectively reduces the inflammatory signals that contribute to chronic conditions such as plaque psoriasis, psoriatic arthritis, and Crohn’s disease. Clinically, this translates into robust efficacy, with significant improvements in disease-specific endpoints and durable responses over long-term treatment periods.

Compared with other therapies, the selective nature of Risankizumab-RZAA not only enhances its efficacy but also mitigates the risk of widespread immunosuppression and adverse events. The favorable safety profile, as reinforced by extensive clinical trial data and ongoing pharmacovigilance, has led to its approval in multiple jurisdictions, affirming its position as a modern and effective therapeutic agent. Furthermore, the ongoing research into diverse indications and optimal dosing strategies underscores its potential to further expand the treatment landscape for various immune-mediated disorders.

Future research directions include an array of ongoing clinical trials that aim to refine patient selection, dosing intervals, and potential combination therapies, as well as explorations into new disease indications where the IL-23/Th17 pathway plays a critical pathological role. Such continued investigation is expected to enhance our understanding of its long-term benefits, ensure continued safety, and possibly broaden its therapeutic application in the years to come.

Overall, Risankizumab-RZAA exemplifies the trend toward precision medicine in immunology, where the deep mechanistic understanding of cytokine biology and cellular signaling is translated into targeted, effective, and relatively safe therapeutic interventions for patients suffering from complex immune-mediated diseases. This paradigm not only offers a significant clinical benefit in terms of symptom relief and disease modification but also holds promise for future discoveries that may lead to further advances in treating chronic inflammatory conditions.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.