1229022-83-6
Enthesitis Related Arthritis (ERA), Juvenile Idiopathic Arthritis (JIA), Juvenile psoriatic arthritis, Non-radiographic Axial Spondyloarthritis, Severe Plaque psoriasis, Active Ankylosing spondylitis, Active Psoriatic arthritis, Moderate Plaque psoriasis
Secukinumab, marketed globally under the brand name Cosentyx®, represents a significant milestone in the management of immune-mediated inflammatory diseases (IMIDs). It is a first-in-class, fully human immunoglobulin G1 kappa (IgG1/κ) monoclonal antibody that selectively targets and neutralizes the proinflammatory cytokine Interleukin-17A (IL-17A).[1] Developed by Novartis, Secukinumab's introduction has fundamentally altered the therapeutic landscape for a spectrum of debilitating chronic conditions, offering a highly targeted mechanism of action that distinguishes it from broader immunosuppressive agents.[1] Its development and approval have validated the IL-17 pathway as a critical therapeutic target in autoimmunity.
The therapeutic rationale for Secukinumab is rooted in its precise inhibition of IL-17A, a pivotal cytokine produced primarily by T helper 17 (Th17) cells.[1] In numerous IMIDs, the IL-23/IL-17 axis is dysregulated, leading to an overproduction of IL-17A, which drives a cascade of inflammatory responses.[4] By binding with high affinity and specificity to IL-17A, Secukinumab prevents the cytokine from interacting with its receptor on various cell types, including keratinocytes and synoviocytes.[1] This action effectively interrupts the downstream signaling that promotes inflammation, tissue damage, and the clinical manifestations of diseases such as psoriasis and spondyloarthropathies.[4] This targeted approach represents a significant evolution from less specific therapies, aiming to maximize efficacy while minimizing off-target effects.
Secukinumab has demonstrated robust, rapid, and sustained efficacy across a progressively expanding list of indications approved by major regulatory bodies like the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA). These indications include moderate-to-severe plaque psoriasis (PsO), active psoriatic arthritis (PsA), active ankylosing spondylitis (AS), active non-radiographic axial spondyloarthritis (nr-axSpA), moderate-to-severe hidradenitis suppurativa (HS), and several pediatric conditions, including enthesitis-related arthritis (ERA) and juvenile psoriatic arthritis (JPsA).[1] An extensive body of clinical trial evidence consistently shows high rates of clinical response, as measured by standard indices such as the Psoriasis Area and Severity Index (PASI 75/90/100), American College of Rheumatology criteria (ACR20/50), and Assessment of SpondyloArthritis international Society criteria (ASAS20/40).[3] In head-to-head trials and long-term studies, its performance has been shown to be superior to placebo and competitive with, or superior to, other established biologics.[3]
The safety profile of Secukinumab is well-characterized through extensive clinical trials and over a decade of real-world use, with more than 5.2 million prescriptions filled in the United States alone.[16] The most common adverse events are mild to moderate upper respiratory tract infections.[2] As a consequence of its mechanism of action, there is an increased risk of certain infections, particularly mucocutaneous candidiasis.[13] Important warnings include the potential for new onset or exacerbation of inflammatory bowel disease (IBD) and the need for pre-treatment screening for tuberculosis.[18] A key differentiator is that Secukinumab does not carry an FDA black box warning, in contrast to some other biologics targeting the IL-17 pathway.[13] This favorable risk-benefit profile, combined with its proven efficacy and convenient dosing, has established Secukinumab as a leading therapeutic choice and a cornerstone treatment for its approved indications.[16]
Secukinumab is a highly specific, engineered biologic therapeutic classified as a recombinant, fully human monoclonal antibody.[1] Its structure conforms to the immunoglobulin G1 kappa (IgG1/κ) isotype, a common framework for therapeutic antibodies known for its long half-life and effector functions.[23] The molecule is a large, symmetrical protein with a molecular weight of approximately 148 to 151 kDa.[4] It is composed of four polypeptide chains: two identical heavy chains, each comprising 457 amino acids, and two identical light chains, each with 215 amino acids.[25] These chains are covalently linked by inter-chain disulfide bonds, forming the characteristic 'Y' shape of an antibody.[27] A critical feature of its structure is the presence of N-linked oligosaccharide chains attached to the heavy chains, a post-translational modification essential for proper protein folding, stability, and function.[26]
Secukinumab is known by the developmental code AIN457 and is identified by the Chemical Abstracts Service (CAS) Number 1229022-83-6 and DrugBank Accession Number DB09029.[1] For clinical use, it is formulated as a sterile, preservative-free liquid solution that is clear to slightly opalescent and colorless to slightly yellow.[20] It is supplied in various single-use formats, including pre-filled syringes, auto-injector pens (Sensoready® and UnoReady®), and a lyophilized powder for reconstitution, which is intended for use by healthcare professionals.[2] In 2023, an intravenous formulation was also approved, expanding its administration options.[2] The formulation is precisely controlled for physicochemical properties such as pH and osmolality to ensure stability and patient tolerability upon injection.[24]
Parameter | Description | Source(s) |
---|---|---|
Generic Name | Secukinumab | 1 |
Brand Name | Cosentyx® | 2 |
DrugBank ID | DB09029 | 1 |
CAS Number | 1229022-83-6 | 24 |
Type | Biotech, Monoclonal Antibody | 1 |
Source | Fully Human | 1 |
Target | Interleukin-17A (IL-17A) | 1 |
Isotype | IgG1/κ | 1 |
Molecular Weight | Approx. 151 kDa | 23 |
Manufacturer | Novartis | 2 |
Table 1: Drug Identification and Physicochemical Properties |
The production of Secukinumab is a complex, multi-stage biopharmaceutical process that underscores the sophistication of modern biotechnology. Unlike small-molecule drugs synthesized through chemical reactions, biologics like Secukinumab are produced by living cells, making the manufacturing process itself a critical determinant of the final product's quality, safety, and efficacy. The process is broadly divided into upstream processing, where the antibody is produced by cells, and downstream processing, where it is purified and formulated.[33] This complexity creates a high barrier to entry for manufacturing and is a primary driver of the cost of biologic therapies, while also explaining the slower development of biosimilars compared to generic small-molecule drugs.
The foundation of Secukinumab manufacturing lies in upstream processing, which begins with cell line development and culminates in the large-scale production of the antibody.[33]
Once the bioreactor culture has reached its peak antibody concentration, the downstream processing phase begins. This phase is designed to harvest, purify, and formulate the antibody to the exceptionally high standards required for a human therapeutic.[33]
Throughout the entire manufacturing process, rigorous quality control testing is performed. A battery of analytical tests is used to monitor the product at every stage, ensuring its identity, purity, potency, and safety. Key quality attributes that are closely monitored include protein concentration, levels of aggregates and fragments, post-translational modifications like glycosylation, endotoxin levels (which must be extremely low, typically <1 EU/mg), and biological activity, which is confirmed using a cell-based assay that measures the antibody's ability to neutralize IL-17A.[24]
The "fully human" designation of Secukinumab is a critical design element with profound clinical implications. It is generated using transgenic mouse technology, where mice are engineered to express the human immunoglobulin gene repertoire instead of their own.[40] When these mice are immunized, they produce fully human antibodies. This approach bypasses the need for "humanization" of a mouse antibody, a process that can still leave some murine sequences that might be recognized as foreign by the human immune system. The direct consequence of this fully human origin is a very low potential for immunogenicity—the formation of anti-drug antibodies (ADAs). Clinical data robustly support this, showing an ADA rate of only 0.4% in large Phase III trials, with no associated loss of efficacy or safety signals.[43] This low immunogenicity is a significant clinical advantage, reducing the risk of treatment failure and hypersensitivity reactions that can plague other biologic therapies and contributing to the drug's long-term viability and reliability for patients.
To understand the mechanism of Secukinumab, it is essential to first appreciate the central role of the Interleukin-17 pathway in the pathogenesis of the diseases it treats. The IL-17 family consists of six related cytokines (IL-17A through IL-17F), but IL-17A is the signature and most potent cytokine of this family, playing a master role in orchestrating chronic inflammation.[1]
IL-17A is produced predominantly by a specialized subset of CD4+ T cells known as T helper 17 (Th17) cells.[1] In healthy individuals, these cells are crucial for host defense, particularly at mucosal surfaces and in clearing fungal and extracellular bacterial infections. However, in a range of IMIDs, this pathway becomes dysregulated. Genetic predisposition and environmental triggers can lead to the over-activation of the IL-23/IL-17 axis.[4] The cytokine IL-23 promotes the expansion and maintenance of pathogenic Th17 cells, which in turn leads to the excessive production and release of IL-17A into tissues and circulation.[7]
Once released, IL-17A exerts its powerful proinflammatory effects by binding to its receptor, the IL-17 receptor (IL-17R), which is widely expressed on numerous cell types, including epithelial cells (like keratinocytes in the skin), endothelial cells, and stromal cells (like fibroblasts and synoviocytes in the joints).[1] The binding of IL-17A to its receptor initiates a cascade of intracellular signaling events, primarily through the activation of key transcription factors such as Nuclear Factor kappa B (NF-κB) and pathways like the mitogen-activated protein kinases (MAPKs).[5]
This signaling cascade results in the transcription and release of a host of other proinflammatory molecules, including:
In the context of specific diseases, this leads to distinct pathologies. In plaque psoriasis, the action of IL-17A on keratinocytes drives their hyperproliferation, leading to the characteristic thickened, red, and scaly plaques (acanthosis and parakeratosis).[1] In
psoriatic arthritis and ankylosing spondylitis, IL-17A promotes synovial inflammation (synovitis), inflammation at the site of tendon and ligament insertions (enthesitis), and drives the processes of bone erosion and pathological new bone formation that lead to joint damage and fusion.[2]
Secukinumab exerts its therapeutic effect through a highly targeted and specific mechanism: the direct neutralization of IL-17A.[1]
As a high-affinity monoclonal antibody, Secukinumab is designed to selectively bind to circulating IL-17A with high precision.[3] This binding action physically prevents IL-17A from docking with its receptor (IL-17R) on target cells.[1] By blocking this crucial first step in the signaling pathway, Secukinumab effectively silences the entire downstream inflammatory cascade that IL-17A would otherwise trigger.[9]
A key feature of Secukinumab's design is its remarkable specificity. It potently neutralizes IL-17A but does not significantly bind to or inhibit other members of the IL-17 family, such as IL-17F.[4] While IL-17F is also proinflammatory, IL-17A is considered the dominant pathogenic driver in these diseases.[6] This deliberate therapeutic strategy of targeting only IL-17A is based on the hypothesis that this focused approach can achieve maximal clinical efficacy while potentially preserving some of the distinct physiological functions of other IL-17 cytokines, which could contribute to a more favorable long-term safety profile compared to broader-acting agents that block the common IL-17 receptor.
The pharmacodynamic effects of this mechanism are profound and measurable. In patients with plaque psoriasis, treatment with Secukinumab leads to a rapid reduction in the clinical signs of the disease—erythema (redness), induration (thickness), and desquamation (scaling)—and a normalization of the skin's histology.[1] In patients with psoriatic arthritis and ankylosing spondylitis, it leads to a marked decrease in systemic inflammatory markers like C-reactive protein (CRP) and tangible improvements in joint swelling, tenderness, and enthesitis.[2] By attenuating the release of proinflammatory cytokines and chemokines, Secukinumab effectively dampens the chronic inflammatory state, providing symptomatic relief and potentially modifying the long-term course of the disease.[1] An interesting pharmacodynamic observation is that total serum IL-17A levels (which include both free IL-17A and the drug-bound complex) increase during treatment. This reflects the formation of stable secukinumab-IL-17A complexes, which have a longer half-life than free IL-17A before they are eventually cleared from the body.[1]
The pharmacokinetic (PK) profile of Secukinumab is characteristic of a human IgG1 monoclonal antibody, defined by slow absorption after subcutaneous injection, limited distribution, slow clearance, and a long elimination half-life. This profile is consistent across its approved indications and provides the scientific basis for its convenient dosing schedules.[4]
Following subcutaneous (SC) administration, Secukinumab is absorbed slowly into the systemic circulation. Peak serum concentrations (Cmax) are typically reached approximately 5 to 6 days post-injection.[1] For a 300 mg dose in plaque psoriasis patients, the mean
Cmax is 27.3 ± 9.5 mcg/mL.[1] The absolute bioavailability of SC-administered Secukinumab is high, estimated to be in the range of 55% to 77%, indicating efficient uptake from the subcutaneous tissue.[1] With the standard maintenance regimen of dosing every 4 weeks, steady-state serum concentrations are achieved by week 24, ensuring consistent therapeutic exposure over the long term.[1] The drug exhibits linear pharmacokinetics, meaning that exposure increases proportionally with the dose, and there is no evidence of dose-dependent clearance.[44]
Secukinumab exhibits a small volume of distribution, indicating that it primarily remains within the vascular and interstitial spaces rather than distributing widely into tissues. The mean volume of distribution during the terminal phase (Vz) has been measured to be between 7.10 and 8.60 L in patients with plaque psoriasis receiving the drug intravenously.[1] This limited distribution is typical for large protein molecules like monoclonal antibodies. Despite this, clinically relevant concentrations are achieved in target tissues. Studies have shown that in psoriatic skin, concentrations of Secukinumab in the interstitial fluid can reach 27% to 40% of the levels found in the serum, allowing it to effectively neutralize IL-17A at the site of inflammation.[6] Body weight has been identified as a significant covariate affecting the drug's PK, with both clearance and volume of distribution increasing in patients with higher body weight.[6]
As a large protein therapeutic, Secukinumab is not metabolized by the hepatic cytochrome P450 (CYP) enzyme system, which is the primary pathway for the metabolism of small-molecule drugs.[4] Instead, it is presumed to be broken down through general protein catabolism pathways. The antibody is likely taken up by cells via endocytosis and degraded into smaller peptides and individual amino acids, which are then recycled by the body.[4] This metabolic route implies that its clearance is not significantly affected by hepatic or renal impairment, simplifying its use in patients with comorbidities affecting these organs.[4]
The systemic clearance (CL) of Secukinumab is low, ranging from 0.14 to 0.22 L/day.[6] This slow clearance is a direct result of its IgG1 structure, which allows it to engage the neonatal Fc receptor (FcRn) pathway. This pathway effectively rescues the antibody from lysosomal degradation and recycles it back into circulation, greatly extending its lifespan in the body. Consequently, Secukinumab has a long mean elimination half-life, estimated to be between 22 and 31 days.[6] This favorable pharmacokinetic property is the cornerstone of the convenient once-monthly maintenance dosing regimen, which enhances patient adherence and quality of life—critical factors in the management of chronic diseases.
The drug interaction profile of Secukinumab is favorable, with few clinically significant interactions identified, primarily due to its metabolic pathway as a large protein.
Secukinumab does not directly inhibit or induce cytochrome P450 enzymes. However, an indirect interaction is possible. The expression and function of certain CYP450 enzymes can be suppressed by elevated levels of proinflammatory cytokines (e.g., IL-1, IL-6, TNF-α) that are characteristic of chronic inflammatory states.[1] By effectively treating the underlying disease and reducing systemic inflammation, Secukinumab can normalize these cytokine levels, which in turn may lead to the "normalization" or increased activity of CYP450 enzymes.[1] This is not a direct drug-drug interaction but rather a drug-disease-drug interaction. This phenomenon could alter the metabolism of co-administered drugs that are CYP450 substrates, particularly those with a narrow therapeutic index such as warfarin or cyclosporine. Therefore, upon initiating or discontinuing Secukinumab in patients taking such medications, monitoring of the therapeutic effect (e.g., INR for warfarin) or drug concentration of the co-administered agent should be considered.[30]
The co-administration of Secukinumab with other biologic agents has not been studied and is generally not recommended due to the potential for additive immunosuppression and a theoretically increased risk of serious infections. In clinical trials for psoriatic arthritis, Secukinumab has been shown to be effective both as monotherapy and when used in combination with methotrexate (MTX), a conventional synthetic disease-modifying antirheumatic drug (csDMARD).[1] No clinically significant pharmacokinetic interactions have been observed between Secukinumab and MTX.
Interacting Agent Class | Specific Examples | Potential Effect | Clinical Recommendation | Source(s) |
---|---|---|---|---|
CYP450 Substrates (Narrow Therapeutic Index) | Warfarin, Cyclosporine, Theophylline | Secukinumab may normalize CYP450 function by reducing inflammation, potentially altering the metabolism and concentration of the substrate drug. | Monitor therapeutic effect or concentration of the substrate drug upon initiation or discontinuation of Secukinumab. | 1 |
Live Vaccines | MMR, Varicella (Chickenpox), Nasal Influenza Vaccine (FluMist), Yellow Fever | Increased risk of disseminated infection from the vaccine virus/bacterium due to immunosuppression. | Contraindicated during treatment. Complete all age-appropriate immunizations prior to initiating Secukinumab. | 28 |
Non-Live (Inactivated) Vaccines | Inactivated Influenza Vaccine, Tdap, COVID-19 Vaccines | The immune response to the vaccine may be reduced, potentially leading to lower vaccine efficacy. | Non-live vaccines can be administered. The potential for a reduced immune response should be considered. | 18 |
Table 8: Key Drug-Drug and Drug-Vaccine Interactions |
Immunogenicity, the propensity of a therapeutic protein to elicit an immune response in the form of anti-drug antibodies (ADAs), is a critical consideration for all biologic therapies. High levels of ADAs, particularly neutralizing antibodies (NAbs), can lead to a loss of drug efficacy by clearing the drug from circulation or blocking its binding site, and can also be associated with hypersensitivity reactions.
Secukinumab has demonstrated a remarkably low immunogenicity profile, a significant clinical and commercial asset that is directly attributable to its fully human antibody structure.[43] In a large, pooled analysis of data from six pivotal Phase III clinical trials in patients with plaque psoriasis, involving 2,842 patients treated with Secukinumab for up to 52 weeks, the incidence of treatment-emergent ADAs (TE-ADAs) was exceptionally low. Only 11 patients (0.4%) developed TE-ADAs during the study period.[43]
Furthermore, of the patients who tested positive for ADAs, only a fraction developed antibodies with neutralizing capacity. Neutralizing antibodies were detected in just three of the nine evaluable patients with TE-ADAs.[43] Most importantly, the presence of either TE-ADAs or neutralizing antibodies was not associated with any discernible impact on the drug's clinical efficacy, its pharmacokinetic profile, or the emergence of any new safety concerns.[43] This very low rate of clinically relevant immunogenicity means that clinicians are less likely to encounter secondary loss of response due to antibody formation, making Secukinumab a more reliable and predictable long-term treatment option compared to biologics with higher rates of ADA development.
The clinical development program for Secukinumab has been extensive, generating a robust body of evidence that supports its efficacy across a wide range of immune-mediated inflammatory diseases. The success of the program has not only established the drug as a standard of care but has also validated the IL-17A pathway as a central driver in these conditions. The progressive expansion of indications reflects a strategic approach, building upon foundational success in psoriasis to address related arthritides and other inflammatory disorders. A notable trend in the clinical trials is the use of high-bar efficacy endpoints, such as PASI 90/100 and complete resolution of enthesitis, signaling a shift in therapeutic goals from mere improvement to achieving disease clearance or minimal disease activity.
Secukinumab's first approval was for moderate-to-severe plaque psoriasis, and its efficacy in this indication is well-established through several large, randomized, placebo- and active-controlled Phase III trials.
Trial Name (Identifier) | Patient Population | Treatment Arms (Dose) | Primary Endpoint | Key Result (Drug vs. Placebo) | p-value | Source(s) |
---|---|---|---|---|---|---|
FIXTURE (NCT01358578) | Adults with moderate-to-severe PsO | Secukinumab 300 mg SC, Secukinumab 150 mg SC, Etanercept 50 mg SC, Placebo | PASI 75 at Week 12 | 77.1% (300 mg) vs. 4.9% | <0.001 | 3 |
ERASURE (NCT01365455) | Adults with moderate-to-severe PsO | Secukinumab 300 mg SC, Secukinumab 150 mg SC, Placebo | PASI 75 at Week 12 | 81.6% (300 mg) vs. 4.5% | <0.001 | 3 |
FEATURE (NCT01555125) | Adults with moderate-to-severe PsO (pre-filled syringe) | Secukinumab 300 mg SC, Secukinumab 150 mg SC, Placebo | PASI 75 at Week 12 | 75.9% (300 mg) vs. 0% | <0.0001 | 3 |
Table 4: Summary of Pivotal Phase III Efficacy Outcomes in Plaque Psoriasis |
Following its success in psoriasis, Secukinumab was developed for psoriatic arthritis, a condition that affects both the skin and joints.
Trial Name (Identifier) | Patient Population | Treatment Arms (Dose) | Primary Endpoint | Key Result (Drug vs. Placebo) | p-value | Source(s) |
---|---|---|---|---|---|---|
FUTURE 2 (NCT01752634) | Adults with active PsA | Secukinumab 300 mg SC, 150 mg SC, 75 mg SC, Placebo | ACR20 at Week 24 | 54% (300 mg) vs. 15% | <0.0001 | 3 |
FUTURE 1 (NCT01392326) | Adults with active PsA | Secukinumab 10 mg/kg IV loading, then 150 mg or 75 mg SC, Placebo | ACR20 at Week 24 | 50.0% (150 mg) vs. 17.3% | <0.001 | 3 |
FUTURE 5 | Adults with active PsA (biologic-naïve and anti-TNF-IR) | Secukinumab 300 mg SC, 150 mg SC, Placebo | ACR20 at Week 16 | 63% (300 mg) vs. 27% | <0.0001 | 13 |
Table 5: Summary of Pivotal Phase III Efficacy Outcomes in Psoriatic Arthritis |
Secukinumab has become a cornerstone therapy for axial spondyloarthropathies, a group of inflammatory diseases primarily affecting the spine and sacroiliac joints.
Trial Name (Identifier) | Patient Population | Treatment Arms (Dose) | Primary Endpoint | Key Result (Drug vs. Placebo) | p-value | Source(s) |
---|---|---|---|---|---|---|
MEASURE 2 (NCT01649375) | Adults with active AS | Secukinumab 150 mg SC, 75 mg SC, Placebo | ASAS20 at Week 16 | 61.1% (150 mg) vs. 28.4% | <0.0001 | 3 |
MEASURE 1 (NCT01358175) | Adults with active AS | Secukinumab IV loading, then 150 mg or 75 mg SC, Placebo | ASAS20 at Week 16 | 60.8% (150 mg) vs. 28.7% | <0.01 | 3 |
INVIGORATE-1 (NCT04156620) | Adults with active axSpA (r-axSpA and nr-axSpA) | Secukinumab IV, Placebo IV | ASAS40 at Week 16 | 40.9% vs. 22.9% | <0.0001 | 14 |
Table 6: Summary of Pivotal Phase III Efficacy Outcomes in Spondyloarthropathies |
In 2023, Secukinumab became the first new biologic therapy approved for moderate-to-severe hidradenitis suppurativa in nearly a decade, addressing a significant unmet need for this chronic, painful, and scarring skin disease.[11]
Secukinumab is approved for active enthesitis-related arthritis in pediatric patients aged 4 years and older.[1] ERA is a category of Juvenile Idiopathic Arthritis (JIA) that is considered a pediatric precursor to adult spondyloarthropathies, sharing a common pathogenic link to the IL-17 pathway. Approval was based on a study demonstrating that children treated with Secukinumab experienced significantly fewer disease flare-ups compared to those randomized to placebo, confirming its efficacy in this younger population.[52]
The safety profile of Secukinumab has been extensively evaluated in a large clinical development program spanning multiple indications and in post-marketing surveillance, establishing it as a generally well-tolerated long-term therapy. The pattern of adverse events is largely predictable based on the drug's mechanism of action—the targeted inhibition of IL-17A.
The overall safety profile is consistent across its approved indications.[46] In long-term observational studies, the incidence of adverse events tends to be highest within the first six months of treatment and then decreases and remains stable over time.[55]
The product label for Secukinumab includes several important warnings and precautions that clinicians and patients must be aware of.
The most significant risk associated with Secukinumab is an increased susceptibility to infections, which is a direct consequence of its mechanism of action.[17] IL-17A plays a vital role in host defense, particularly in maintaining mucosal barrier integrity and orchestrating the immune response against fungal and certain bacterial pathogens.[5]
The role of IL-17 in the gut is complex and context-dependent; while it can be pathogenic, it can also play a protective role in maintaining gut mucosal integrity. Blocking IL-17A can disrupt this balance.
The only absolute contraindication for Secukinumab is a history of a previous serious hypersensitivity reaction to the active substance, secukinumab, or to any of the excipients in the formulation.[13]
A significant feature of Secukinumab's safety profile is the absence of an FDA-issued "black box warning".[13] The FDA reserves this type of warning for drugs with the most serious or life-threatening risks. This "non-finding" is a powerful differentiator that shapes clinical perception and prescribing behavior. It contrasts sharply with another drug in the IL-17 inhibitor class, the IL-17
receptor antagonist brodalumab (Siliq), which carries a black box warning for suicidal ideation and behavior observed in its clinical trials.[22] Although a causal relationship for brodalumab was not definitively established, the warning imposes a significant clinical and administrative burden, including a restricted access program (REMS). The absence of such a warning for Secukinumab gives it a substantial advantage, making it a more straightforward choice for clinicians, particularly when treating patients with a history of depression or other mental health conditions.
System Organ Class | Adverse Event | Frequency | Clinical Context/Comments | Source(s) |
---|---|---|---|---|
Infections and Infestations | Upper Respiratory Tract Infection (Nasopharyngitis) | Very Common (>10%) | Most frequent AE; typically mild to moderate. | 2 |
Mucocutaneous Candidiasis (Oral, Esophageal, Vulvovaginal) | Common (1-10%) | Dose-dependent risk. Consistent with MOA. Usually manageable with standard antifungals. | 13 | |
Herpes Simplex (Oral) | Common (1-10%) | Reactivation of latent virus. | 2 | |
Tuberculosis (Reactivation) | Rare (<1%) | Pre-treatment screening for latent/active TB is mandatory. | 20 | |
Gastrointestinal Disorders | Diarrhea | Common (1-10%) | Usually mild and transient. | 2 |
Inflammatory Bowel Disease (New Onset or Exacerbation) | Uncommon (<1%) | A key warning. Caution in patients with a history of IBD. Monitor for new/worsening symptoms. | 18 | |
Immune System Disorders | Hypersensitivity Reactions (Urticaria, Anaphylaxis) | Rare (<1%) | Anaphylaxis is a contraindication to future use. Discontinue immediately if serious reaction occurs. | 18 |
Skin and Subcutaneous Tissue Disorders | Injection Site Reactions | Common (1-10%) | Typically mild erythema, pruritus, or swelling. | 2 |
Hepatobiliary Disorders | Hepatitis B Reactivation | Rare (<1%) | Can occur in susceptible carriers. Screening and potential prophylaxis are recommended. | 46 |
Table 7: Comprehensive Safety Profile and Adverse Event Frequencies |
The dosing and administration of Secukinumab are highly specific and tailored to the indication, patient age, and in some cases, body weight. The availability of multiple formulations and dosing strategies provides flexibility for clinicians to optimize treatment for individual patients.
Secukinumab regimens typically consist of an initial weekly "loading" phase to rapidly achieve therapeutic concentrations, followed by a less frequent "maintenance" phase.
Indication (Population) | Route | Loading Dose | Maintenance Dose | Key Notes |
---|---|---|---|---|
Plaque Psoriasis (Adult) | SC | 300 mg at Weeks 0, 1, 2, 3, 4 | 300 mg every 4 weeks | 150 mg dose may be acceptable for some. |
Plaque Psoriasis (Pediatric, ≥6 yrs) | SC | Weeks 0, 1, 2, 3, 4 | Every 4 weeks | Weight-based: <50 kg: 75 mg; ≥50 kg: 150 mg. |
Psoriatic Arthritis (Adult) | SC | Optional: 150 mg at Weeks 0, 1, 2, 3, 4 | 150 mg every 4 weeks | Can increase to 300 mg. Use 300 mg regimen for coexistent mod-sev PsO. |
IV | Optional: 6 mg/kg at Week 0 | 1.75 mg/kg every 4 weeks | Administered over 30 min by HCP. | |
Ankylosing Spondylitis (Adult) | SC | Optional: 150 mg at Weeks 0, 1, 2, 3, 4 | 150 mg every 4 weeks | Can increase to 300 mg if response is inadequate. |
IV | Optional: 6 mg/kg at Week 0 | 1.75 mg/kg every 4 weeks | Administered over 30 min by HCP. | |
nr-axSpA (Adult) | SC | Optional: 150 mg at Weeks 0, 1, 2, 3, 4 | 150 mg every 4 weeks | - |
IV | Optional: 6 mg/kg at Week 0 | 1.75 mg/kg every 4 weeks | Administered over 30 min by HCP. | |
Hidradenitis Suppurativa (Adult) | SC | 300 mg at Weeks 0, 1, 2, 3, 4 | 300 mg every 4 weeks | Can increase frequency to every 2 weeks if needed. |
JPsA (≥2 yrs) & ERA (≥4 yrs) | SC | Weeks 0, 1, 2, 3, 4 | Every 4 weeks | Weight-based: 15-<50 kg: 75 mg; ≥50 kg: 150 mg. |
Table 3: Dosing and Administration Regimens by Indication |
Proper administration technique is crucial for ensuring efficacy and minimizing local reactions.
Secukinumab was discovered, developed, and is manufactured by the global pharmaceutical company Novartis.[2] During its development phase, it was identified by the research code AIN457.[3] It is marketed worldwide under the proprietary brand name Cosentyx®.[1]
The regulatory journey of Secukinumab showcases a highly successful and strategic clinical development program, marked by a steady expansion of approved indications following its initial launch. This phased approach allowed Novartis to build a massive evidence base, gain clinician familiarity, and systematically broaden the drug's market.
The FDA approval timeline demonstrates a logical progression from a large primary indication to related diseases and special populations:
The EMA's approval history has largely paralleled the FDA's, establishing Secukinumab as a key therapy across Europe:
Indication | Patient Population | Approving Agency | Date of First Approval for Indication |
---|---|---|---|
Plaque Psoriasis | Adults (moderate-to-severe) | FDA / EMA | Jan 2015 |
Psoriatic Arthritis | Adults (active) | FDA / EMA | Jan 2016 |
Ankylosing Spondylitis | Adults (active) | FDA / EMA | Jan 2016 |
Non-Radiographic Axial Spondyloarthritis | Adults (active, with objective signs of inflammation) | FDA / EMA | Jun 2020 (FDA) |
Plaque Psoriasis (Pediatric) | ≥6 years (moderate-to-severe) | FDA / EMA | Jun 2021 (FDA) |
Enthesitis-Related Arthritis (Pediatric) | ≥4 years (active) | FDA / EMA | Dec 2021 (FDA) |
Juvenile Psoriatic Arthritis (Pediatric) | ≥2 years (active) | FDA / EMA | Dec 2021 (FDA) |
Hidradenitis Suppurativa | Adults (moderate-to-severe) | EMA / FDA | Jun 2023 (EMA) / Oct 2023 (FDA) |
Intravenous Formulation | Adults (PsA, AS, nr-axSpA) | FDA | Oct 2023 |
Table 2: Summary of Key FDA and EMA Approved Indications |
As the first-in-class IL-17A inhibitor, Secukinumab enjoyed a significant first-to-market advantage, allowing it to establish a strong foothold with clinicians and patients.[2] Its commercial success has been substantial, with over 5.2 million prescriptions filled in the U.S. by late 2024, making it a blockbuster product for Novartis.[16]
The success of Secukinumab has also validated the IL-17 pathway as a highly lucrative therapeutic target, which has spurred intense competition. The market now includes several other biologics that target the same pathway, creating a highly competitive landscape. Key competitors include:
In this crowded market, differentiation is based on multiple factors, including efficacy (speed of onset, depth of response, durability), safety (particularly the IBD risk and the presence or absence of black box warnings), dosing convenience (subcutaneous vs. intravenous, dosing frequency), and the breadth of approved indications. Secukinumab's continued investment in clinical trials and indication expansion is a core strategy to maintain its competitive leadership.
Secukinumab (Cosentyx®) has established itself as a transformative therapy in the field of immunology. As the first-in-class selective IL-17A inhibitor, it offers a highly targeted mechanism of action that has proven to be exceptionally effective across a broad and expanding range of chronic immune-mediated inflammatory diseases. Its molecular design as a fully human monoclonal antibody translates into a clinically significant low immunogenicity profile, contributing to its reliable and durable long-term efficacy.
The extensive clinical trial program has consistently demonstrated rapid and profound improvements in the signs and symptoms of plaque psoriasis, psoriatic arthritis, axial spondyloarthropathies, and hidradenitis suppurativa, setting new benchmarks for therapeutic success in these conditions. The drug's safety profile is well-characterized and considered favorable, with the most common adverse events being a predictable consequence of its targeted immunomodulation. The absence of an FDA black box warning further strengthens its risk-benefit proposition for clinicians and patients.
Supported by a favorable pharmacokinetic profile that allows for convenient monthly maintenance dosing and a strategic expansion into pediatric populations and new formulations, Secukinumab stands as a cornerstone of modern therapy for IL-17A-driven diseases. Its journey from a novel molecular target to a multi-indication blockbuster therapeutic exemplifies the success of rational drug design and targeted immunotherapy in modern medicine.
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.