Stelara, Pyzchiva, Uzpruvo, Wezenla, Steqeyma, Otulfi, Fymskina, Eksunbi, Absimky, Imuldosa, Yesintek, Qoyvolma, Usymro
Biotech
815610-63-0
Severe Plaque psoriasis, Ulcerative Colitis, Active Severe, Active Psoriatic arthritis, Moderate Plaque psoriasis, Moderate Ulcerative colitis, Moderate, active Crohn´s Disease, Severe, active Crohn´s Disease
Ustekinumab is a first-in-class, fully human IgG1κ monoclonal antibody that represents a significant milestone in the treatment of immune-mediated inflammatory diseases. Marketed under the brand name Stelara® and now available as multiple biosimilars, it functions as a dual inhibitor, uniquely targeting the shared p40 protein subunit of two critical cytokines: Interleukin-12 (IL-12) and Interleukin-23 (IL-23).[1] This targeted mechanism of action allows Ustekinumab to potently downregulate the Th1 and Th17 inflammatory pathways, which are central to the pathophysiology of several autoimmune conditions.[1]
The therapeutic utility of Ustekinumab has been firmly established through a robust clinical development program, leading to regulatory approvals for a range of indications. These include the treatment of moderate-to-severe plaque psoriasis (PsO), active psoriatic arthritis (PsA), moderately to severely active Crohn's disease (CD), and moderately to severely active ulcerative colitis (UC).[1] Its approval extends to pediatric populations for both plaque psoriasis and psoriatic arthritis, underscoring its well-characterized efficacy and safety profile across different age groups.[7] Clinical trials have consistently demonstrated that Ustekinumab can achieve significant clinical endpoints, including skin clearance in psoriasis, joint symptom improvement in psoriatic arthritis, and clinical and endoscopic remission in inflammatory bowel diseases.[9]
The safety profile of Ustekinumab is well-defined and is a direct consequence of its immunomodulatory mechanism. While generally well-tolerated, its primary risks include an increased susceptibility to infections, necessitating pre-treatment screening for tuberculosis and vigilance for other serious infections during therapy.[1] Its long pharmacokinetic half-life of approximately three weeks supports convenient maintenance dosing regimens of every 8 or 12 weeks, a significant advantage for patient adherence.[1]
As a blockbuster therapeutic, Ustekinumab has had a profound impact on clinical practice and the pharmaceutical market. The recent market entry of multiple FDA- and EMA-approved biosimilars marks a pivotal moment, poised to dramatically alter the commercial landscape by increasing competition and expanding patient access through lower costs.[15] The evolution of Ustekinumab, from its initial approval for psoriasis to its current role as a multi-indication therapy and its ongoing investigation for new uses, highlights its enduring importance in the armamentarium against chronic inflammatory diseases.
This section establishes the fundamental identity of Ustekinumab, providing a definitive reference for its biological classification, nomenclature, and key physicochemical properties.
Ustekinumab is classified as a biotech drug, specifically a fully human immunoglobulin G1 kappa (IgG1κ) monoclonal antibody.[1] Within the therapeutic landscape, it is categorized as an interleukin inhibitor, a biologic Disease-Modifying Anti-Rheumatic Drug (bDMARD), and a systemic antipsoriatic agent.[5] Its development represents a significant advancement in targeted immunotherapy. The antibody was generated through the immunization of human immunoglobulin (hu-Ig) transgenic mice with recombinant human IL-12, a process that yields a fully human antibody, thereby minimizing the potential for immunogenicity that can be associated with chimeric or humanized antibodies.[5] The development was pioneered by Centocor Ortho Biotech, which later became part of Janssen, a pharmaceutical company of Johnson & Johnson.[1]
To ensure unambiguous identification in clinical, research, and regulatory contexts, Ustekinumab is associated with a range of standard identifiers and commercial names. The reference product is globally recognized as Stelara®. Following the expiration of key patents, a number of biosimilar products have been approved and launched, particularly in the United States and European Union.
The U.S. Food and Drug Administration (FDA) has adopted a specific naming convention for biologics, appending a unique, meaningless four-letter suffix to the nonproprietary name of each biosimilar product. This system is designed to enhance pharmacovigilance by enabling precise tracking of adverse events to a specific manufacturer's product and to prevent the inadvertent substitution of products that have not been designated as "interchangeable".[15] In contrast, the European Medicines Agency (EMA) approves biosimilars with distinct brand names but does not utilize the suffix system for the nonproprietary name, a key difference in global regulatory practice.[17] The consolidation of these various names and identifiers is crucial for healthcare professionals, researchers, and industry analysts.
Table 1: Ustekinumab Identifiers and Commercial Names
Category | Identifier/Name | Source(s) |
---|---|---|
Generic Name | Ustekinumab | 5 |
DrugBank ID | DB05679 | 1 |
CAS Number | 815610-63-0 | 1 |
ATC Code | L04AC05 | 1 |
Other Identifiers | UNII: FU77B4U5Z0KEGG: D09214ChEMBL: ChEMBL1201835 | 1 |
Developmental Code | CNTO 1275, CNTO-1275 | 21 |
Reference Product | Stelara® | 1 |
Approved Biosimilars | Wezlana (ustekinumab-auub)Selarsdi (ustekinumab-aekn)Pyzchiva (ustekinumab-ttwe)Otulfi (ustekinumab-aauz)Yesintek (ustekinumab-kfce)Imuldosa (ustekinumab-srlf)Steqeyma (ustekinumab-stba)Starjemza (ustekinumab-hmny)Uzpruvo (EU) | 15 |
As a large protein therapeutic, Ustekinumab possesses physicochemical properties characteristic of a monoclonal antibody.
The therapeutic efficacy of Ustekinumab is derived from its precise and targeted interference with the Interleukin-12 (IL-12) and Interleukin-23 (IL-23) signaling pathways. This section provides a detailed scientific explanation of its mechanism, from the broader immunological context to the specific molecular interactions that define its function.
IL-12 and IL-23 are naturally occurring, heterodimeric cytokines that serve as master regulators of adaptive immunity, but their dysregulation is a cornerstone of the pathophysiology of many autoimmune diseases.[5] Both cytokines are produced by antigen-presenting cells, such as dendritic cells and macrophages, and are found at elevated levels in the affected tissues—including the skin, joints, and gastrointestinal mucosa—of patients with psoriasis, psoriatic arthritis, and inflammatory bowel disease.[2]
The IL-12 pathway is central to the development of T-helper 1 (Th1) cell-mediated immunity. IL-12, which is composed of a p35 and a p40 subunit, promotes the differentiation of naive T-cells into Th1 cells. These Th1 cells are crucial for defending against intracellular pathogens but, when overactivated in autoimmune states, they produce high levels of pro-inflammatory cytokines, most notably interferon-gamma (IFNγ), which drives tissue inflammation and damage.[18]
The IL-23 pathway is essential for the maintenance and expansion of the T-helper 17 (Th17) cell lineage. IL-23 is composed of a p19 and a p40 subunit. Its primary role is to sustain Th17 cells, which are potent producers of inflammatory cytokines like IL-17A, IL-17F, and IL-22. The Th17 pathway is now understood to be a dominant driver of pathology in conditions like psoriasis and has a critical role in the chronic inflammation characteristic of psoriatic arthritis and inflammatory bowel disease.[1]
A fascinating aspect of Ustekinumab's development history is that its dual specificity was a serendipitous discovery rather than an initial design feature. The drug was developed to target the p40 subunit, which was known to be part of IL-12. The existence and pro-inflammatory role of IL-23, which also utilizes the p40 subunit, were described after the initial discovery and preclinical development of Ustekinumab.[18] This realization transformed the understanding of the drug's mechanism, elevating it from a specific IL-12 inhibitor to a broader immunomodulator that could simultaneously suppress both the Th1 and the highly pathogenic Th17 pathways. This "accidental" dual targeting is a key reason for its broad efficacy across multiple distinct autoimmune diseases.
The p40 protein is the common structural element shared by both IL-12 and IL-23, making it an ideal strategic target for simultaneously neutralizing both cytokines.[18] Ustekinumab is a high-affinity human IgG1κ monoclonal antibody engineered to bind with exceptional specificity to this p40 subunit.[1] This binding is a direct, physical interaction confirmed by co-crystal structure analysis, where the fragment antigen-binding (Fab) portion of the antibody engages the p40 protein.[18] This action effectively sequesters the p40 subunit, preventing its assembly into functional IL-12 or IL-23 heterodimers and blocking their ability to interact with their respective cell surface receptors.
The therapeutic effect of Ustekinumab is realized through the downstream consequences of its p40 blockade. By binding to p40, Ustekinumab prevents both IL-12 and IL-23 from docking with their shared receptor component, the IL-12 receptor beta 1 (IL-12Rβ1) chain, which is expressed on the surface of immune effector cells like natural killer (NK) cells and T-cells.[1]
This receptor blockade prevents the initiation of intracellular signaling cascades. For the IL-12 pathway, it inhibits the phosphorylation of the signal transducer and activator of transcription 4 (STAT4). For the IL-23 pathway, it blocks the phosphorylation of STAT3.[18] The interruption of these critical signaling events has profound functional consequences:
By disrupting these cascades, Ustekinumab effectively dampens the overactive immune response that drives chronic inflammation in its target diseases. This mechanism, while highly effective, also provided the scientific rationale for the development of its successors. The success of Ustekinumab validated the IL-12/23 axis as a therapeutic target. This led to the development of second-generation biologics that are more selective, targeting only the p19 subunit of IL-23 (e.g., risankizumab, guselkumab).[9] The hypothesis behind this evolution is that specifically inhibiting the Th17 pathway via IL-23p19 blockade may offer comparable or superior efficacy, particularly in diseases like psoriasis, while potentially improving the safety profile by sparing the IL-12/Th1 pathway, which plays a role in host defense. Thus, Ustekinumab's mechanism is both its core strength and the foundation upon which its competition was built.
The clinical use of Ustekinumab is guided by its distinct pharmacokinetic (PK) and pharmacodynamic (PD) properties. Its absorption, distribution, metabolism, and elimination profile dictates its dosing regimen, while its pharmacodynamic effects correlate directly with its therapeutic mechanism.
The pharmacokinetic profile of Ustekinumab is characterized by slow absorption after subcutaneous administration, limited distribution, a long elimination half-life, and low clearance, all typical features of a monoclonal antibody therapeutic.
Table 2: Summary of Key Pharmacokinetic Parameters for Ustekinumab
Parameter | Value | Patient Population / Notes | Source(s) |
---|---|---|---|
Route of Administration | Subcutaneous (SC), Intravenous (IV) | SC for maintenance; IV for IBD induction | 1 |
Bioavailability (F) | 57.2% | After single SC dose | 5 |
Time to Max Concentration (Tmax) | 7 - 13.5 days | After single SC dose in PsO patients | 5 |
Volume of Distribution (Vd) | 4.4 - 4.6 L | Steady-state in IBD patients | 5 |
Metabolism | Catabolic pathways (protein degradation) | Not metabolized by CYP450 enzymes | 5 |
Elimination Half-Life (t1/2) | ~19 - 21 days (average 3 weeks) | Range: 15-32 days across studies | 1 |
Clearance (CL) | ~0.19 L/day | In IBD patients | 5 |
The pharmacodynamic effects of Ustekinumab are a direct manifestation of its mechanism of action. By binding the p40 subunit and blocking IL-12 and IL-23 signaling, Ustekinumab produces measurable changes in the inflammatory milieu.[5] Treatment leads to a significant downregulation in the gene expression and serum levels of various pro-inflammatory cytokines and chemokines that are downstream of the IL-12/23 axis. These include reductions in mediators such as Monocyte Chemoattractant Protein-1 (MCP-1), Tumor Necrosis Factor-alpha (TNF-α), Interferon-inducible Protein-10 (IP-10), and IL-8.[5] The suppression of these biomarkers of inflammation provides a biological measure of the drug's activity and correlates with the clinical improvements observed in patients.
Ustekinumab has undergone extensive clinical investigation, leading to its approval by major regulatory bodies like the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for several distinct immune-mediated inflammatory diseases. This section details the pivotal clinical trial evidence supporting its use for each approved indication. The chronological expansion of these indications—from plaque psoriasis in 2009, to psoriatic arthritis in 2013, Crohn's disease in 2016, and ulcerative colitis in 2019—illustrates a highly successful lifecycle management strategy.[20] This "pipeline in a product" approach strategically leveraged the understanding of a common underlying pathophysiology across different medical specialties, transforming a single drug into a multi-specialty cornerstone therapy.
Table 3: Efficacy in Pivotal Adult Plaque Psoriasis Trials
Trial / Comparison | Treatment Arms | Primary Endpoint | Result at Week 12 | Source(s) |
---|---|---|---|---|
PHOENIX 1 & 2 | Ustekinumab 45 mg & 90 mgPlacebo | % of patients with PASI 75 response | ~69% (Ustekinumab)~3% (Placebo) | 12 |
ACCEPT | Ustekinumab 45 mg & 90 mgEtanercept 50 mg | % of patients with PASI 75 response | Ustekinumab was significantly more effective than Etanercept | 9 |
Table 4: Efficacy in Pivotal Psoriatic Arthritis Trials (ACR20 Response at Week 24)
Trial | Treatment Arms | % of Patients with ACR20 Response | Source(s) |
---|---|---|---|
PSUMMIT I | Ustekinumab 45 mgUstekinumab 90 mgPlacebo | 42%50%23% | 11 |
PSUMMIT II | Ustekinumab 45 mgUstekinumab 90 mgPlacebo | 44%44%20% | 11 |
The practical application of Ustekinumab in clinical practice requires a clear understanding of its available formulations and the specific, often complex, dosing regimens tailored to each indication and patient population.
Ustekinumab is supplied in sterile, preservative-free liquid solutions for parenteral administration. The available formats are designed to accommodate both intravenous loading doses and subcutaneous maintenance therapy.[14]
The dosing strategy for Ustekinumab varies significantly depending on the disease being treated, the patient's body weight, and whether the treatment is for induction or maintenance. A particularly important distinction is the bimodal administration strategy for inflammatory bowel disease (IBD). This approach utilizes a one-time, weight-based IV induction dose to rapidly achieve therapeutic drug concentrations and induce a swift clinical response in acutely ill patients. This is followed by a transition to a simpler, fixed-dose SC maintenance regimen for long-term management. This strategy is tailored to the distinct clinical needs of IBD patients, where rapid control of severe inflammation is paramount, compared to the all-subcutaneous regimen used for psoriatic diseases.
Furthermore, the emergence of biosimilars has introduced a critical nuance for pediatric care. Not all biosimilar manufacturers have developed formulations that permit the precise weight-based dosing required for children weighing less than 60 kg. For instance, some biosimilars may not be suitable for this patient group, requiring clinicians to select the originator product or a biosimilar that specifically offers the appropriate dosage forms.[15] Conversely, some biosimilar developers are actively seeking approvals for new pediatric-friendly formulations to address this gap, creating a dynamic and fragmented market where product choice can be dictated by patient weight.[16]
Table 7: Dosing and Administration Regimens for Ustekinumab by Indication
Indication | Patient Population | Induction Dose | Maintenance Dose | Source(s) |
---|---|---|---|---|
Plaque Psoriasis (PsO) | Adults | ≤100 kg: 45 mg SC at weeks 0 & 4>100 kg: 90 mg SC at weeks 0 & 4 | ≤100 kg: 45 mg SC every 12 weeks>100 kg: 90 mg SC every 12 weeks | 7 |
Pediatric (≥6 yrs) | <60 kg: 0.75 mg/kg SC at weeks 0 & 460-100 kg: 45 mg SC at weeks 0 & 4>100 kg: 90 mg SC at weeks 0 & 4 | <60 kg: 0.75 mg/kg SC every 12 weeks60-100 kg: 45 mg SC every 12 weeks>100 kg: 90 mg SC every 12 weeks | 7 | |
Psoriatic Arthritis (PsA) | Adults | 45 mg SC at weeks 0 & 4(90 mg if >100 kg with co-existent mod-sev PsO) | 45 mg SC every 12 weeks(90 mg if >100 kg with co-existent mod-sev PsO) | 7 |
Pediatric (≥6 yrs) | <60 kg: 0.75 mg/kg SC at weeks 0 & 4≥60 kg: 45 mg SC at weeks 0 & 4(90 mg if >100 kg with co-existent mod-sev PsO) | <60 kg: 0.75 mg/kg SC every 12 weeks≥60 kg: 45 mg SC every 12 weeks(90 mg if >100 kg with co-existent mod-sev PsO) | 7 | |
Crohn's Disease (CD) & Ulcerative Colitis (UC) | Adults | Single IV infusion based on weight:≤55 kg: 260 mg>55 kg to ≤85 kg: 390 mg>85 kg: 520 mg | 90 mg SC 8 weeks after IV dose, then 90 mg SC every 8 weeks | 7 |
The safety profile of Ustekinumab has been extensively characterized through a large body of clinical trial data and over a decade of post-marketing experience. While generally well-tolerated, its use is associated with specific risks that are a direct and predictable consequence of its immunomodulatory mechanism of action. Suppressing the Th1 and Th17 pathways, which are essential for host defense against certain types of pathogens, logically leads to an increased risk of infections.
The prescribing information for Ustekinumab and its biosimilars includes several key warnings and precautions to guide safe use.
The use of Ustekinumab is contraindicated in patients with a known history of clinically significant hypersensitivity to the active substance or to any of the excipients in the formulation.[6] It is also contraindicated in patients with a clinically important, active infection.[14]
The potential for drug-drug interactions with Ustekinumab is relatively low, primarily due to its specific mechanism of action and metabolic pathway. However, certain combinations, particularly with other immunomodulating agents, require careful consideration.
The concomitant use of Ustekinumab with other biologic therapies that suppress the immune system is not recommended. Combining Ustekinumab with TNF antagonists (e.g., adalimumab, infliximab, etanercept) or other biologics such as abatacept, anakinra, or rituximab has not been studied in depth but is expected to increase the risk of serious infections without providing additional clinical benefit.[30] Therefore, such combinations should be avoided.
As detailed in the Clinical Pharmacology section, Ustekinumab is a monoclonal antibody that is degraded via catabolic pathways and is not metabolized by the cytochrome P450 (CYP) enzyme system.[5] Clinical studies have confirmed that it does not have a clinically significant effect on the activity of major CYP enzymes.[5] This is a key advantage, as it means that Ustekinumab is not expected to alter the metabolism of co-administered drugs that are substrates for these enzymes. Consequently, no dose adjustments are required for concomitant medications such as warfarin, cyclosporine, or others that are metabolized through CYP pathways.[8] This low potential for metabolic interactions simplifies its use in patients with multiple comorbidities who are often on complex medication regimens.
The interaction between Ustekinumab and allergen immunotherapy (allergy shots) has not been formally studied. Ustekinumab works by modulating the immune system, and it is theoretically possible that it could interfere with the efficacy of allergen immunotherapy or, conversely, increase the risk of allergic reactions associated with it. Caution is advised if these treatments are used concurrently.[7]
Ustekinumab's journey from a novel investigational compound to a multi-billion-dollar blockbuster therapeutic, and now to a market facing biosimilar competition, provides a compelling case study in modern pharmaceutical development, lifecycle management, and market dynamics.
Ustekinumab was developed by Centocor, a subsidiary of Johnson & Johnson, with foundational technology from Medarex, and is marketed globally by Janssen Pharmaceuticals.[1] Its regulatory path was marked by a strategic, indication-by-indication expansion that leveraged a growing understanding of the central role of the IL-12/23 axis in various autoimmune diseases. This methodical approach allowed the drug to penetrate multiple medical specialties, including dermatology, rheumatology, and gastroenterology. The timeline of its major regulatory milestones in the US and Europe highlights this successful strategy.
Table 9: Key FDA and EMA Approval Timeline for Stelara® (Ustekinumab)
Date | Regulatory Body | Action / Indication | Source(s) |
---|---|---|---|
Dec 2007 | FDA / EMA | Marketing applications submitted for moderate to severe plaque psoriasis | 20 |
Sep 25, 2009 | FDA | Approval for moderate to severe Plaque Psoriasis (Adults) | 20 |
Sep 23, 2013 | FDA | Approval for active Psoriatic Arthritis (Adults) | 11 |
Nov 11, 2016 | EMA | Approval for moderately to severely active Crohn's Disease (Adults) | 26 |
Sep 26, 2016 | FDA | Approval for moderately to severely active Crohn's Disease (Adults) | 20 |
Oct 13, 2017 | FDA | Approval for moderate to severe Plaque Psoriasis (Adolescents) | 20 |
Oct 21, 2019 | FDA | Approval for moderately to severely active Ulcerative Colitis (Adults) | 20 |
Jul 30, 2020 | FDA | Approval for moderate to severe Plaque Psoriasis (Pediatric ≥6 yrs) | 20 |
Aug 1, 2022 | FDA | Approval for active Psoriatic Arthritis (Pediatric ≥6 yrs) | 20 |
Apr 2025 (est.) | EMA | Approval for moderately to severely active Crohn's Disease (Pediatric) | 27 |
For many years, Stelara® enjoyed market exclusivity, generating billions of dollars in annual revenue for Johnson & Johnson. The expiration of its primary patents and supplementary protection certificates in the mid-2020s has ushered in a new era of competition from biosimilar products.[16] A biosimilar is a biological product that is highly similar to and has no clinically meaningful differences from an existing FDA-approved reference product.[7]
The entry of Ustekinumab biosimilars has not been a single event but rather a "wave" of launches staggered throughout 2025. This pattern is the result of complex legal negotiations and settlement agreements—often referred to as the "patent dance"—between the innovator company (J&J) and each individual biosimilar manufacturer. These agreements dictate the precise date on which each competitor can enter the market, creating a dynamic and complex transition period.[16]
As of early 2025, several biosimilars have been approved by the FDA and/or EMA and have begun to launch, including:
[15]
A key regulatory distinction in the US market is the concept of interchangeability. An interchangeable biosimilar has met additional FDA requirements demonstrating that it can be expected to produce the same clinical result as the reference product in any given patient and that the risk of switching between the two is no greater than using the reference product alone. This designation allows for substitution by a pharmacist without the intervention of the prescribing physician, potentially accelerating uptake.[15]
The implications of this market shift are profound. The introduction of multiple biosimilars is expected to drive significant price competition, leading to substantial cost savings for healthcare systems and payers. This, in turn, is anticipated to improve patient access to this important class of therapy. For clinicians and health systems, this new landscape will require careful management of formularies and prescribing choices, balancing factors of cost, brand familiarity, and specific product attributes like interchangeability status or the availability of pediatric-friendly formulations.
While Ustekinumab is a mature product with a well-established role in several major inflammatory diseases, research continues to explore its potential utility in other conditions and specific patient populations. This ongoing investigation represents a strategy to further define the therapeutic boundaries of IL-12/23 inhibition and potentially expand the drug's label, a key consideration in the face of increasing biosimilar competition.
Based on the shared inflammatory pathways involving IL-12 and IL-23, Ustekinumab has been used off-label by clinicians to treat a variety of other autoimmune and inflammatory conditions where these cytokines are implicated. Documented off-label uses include hidradenitis suppurativa, Behçet disease, pyoderma gangrenosum, and certain forms of arteritis, although the evidence for these uses is less robust than for its approved indications.[23]
The clinical trial landscape for Ustekinumab remains active, with studies focused on both expanding into new therapeutic areas and generating more data within currently approved indications. These trials serve as a roadmap for the drug's future, representing strategic efforts to either find new markets or solidify its position in existing ones.
Key investigational areas include:
Ustekinumab holds a pivotal place in the history of immunology and therapeutic medicine. As the first agent to successfully target the IL-12/23 pathway, it not only provided a powerful new treatment option for millions of patients but also validated a novel biological target, paving the way for a subsequent generation of more selective therapies. This concluding analysis synthesizes its clinical positioning, key considerations for practice, and its likely future trajectory in an evolving healthcare landscape.
Ustekinumab occupies a unique and important niche in the treatment algorithms for chronic inflammatory diseases. Its position relative to other biologic classes is nuanced and indication-specific.
Compared to its direct competitors, Ustekinumab sits in a unique mechanistic space. It offers a more targeted approach than the broad immunosuppression of TNF-alpha inhibitors. At the same time, it is less selective than the newer IL-23p19 inhibitors (e.g., risankizumab, guselkumab) and IL-17 inhibitors (e.g., secukinumab, ixekizumab). This dual IL-12/23 inhibition is both its defining feature and a point of clinical debate. While some argue that the added IL-12 blockade may contribute to certain infection risks without adding significant efficacy in some diseases, others value its long-established track record and efficacy across a broad spectrum of conditions, from skin to joints to gut.
For practicing clinicians, the effective use of Ustekinumab requires several key considerations:
The future of Ustekinumab will be shaped by two opposing forces: the erosion of its market share by lower-cost biosimilars and the enduring value of its decade-plus legacy of proven safety and efficacy. It will likely remain a cornerstone therapy for the foreseeable future, particularly as biosimilar competition drives down its cost and increases its accessibility. However, its role as a first-choice agent may be challenged in some areas, especially psoriasis, by the newer, more selective IL-23 and IL-17 inhibitors as they accumulate more long-term data.
Ultimately, Ustekinumab's value proposition will increasingly hinge on a compelling balance between cost-effectiveness (driven by the biosimilar market) and the clinical confidence that comes with a well-understood, reliable, and broadly effective therapy. Its story is a testament to the power of targeted immunology and a paradigm for successful drug development in the modern era.
Published at: July 11, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.