MedPath

Tocilizumab Advanced Drug Monograph

Published:Jul 10, 2025

Generic Name

Tocilizumab

Brand Names

Actemra, RoActemra, Tyenne, Tofidence, Avtozma

Drug Type

Biotech

CAS Number

375823-41-9

Associated Conditions

Coronavirus Disease 2019 (COVID‑19), Cytokine Release Syndrome caused by CAR-T Cell Therapy, Giant Cell Arteritis (GCA), Juvenile Chronic Polyarthritis, Moderate to Severe Rheumatoid Arthritis, Moderately to Severely Active Rheumatoid Arthritis, Polyarticular Juvenile Idiopathic Arthritis, Systemic Juvenile Idiopathic Arthritis (SJIA), Systemic Sclerosis-associated Interstitial Lung Disease (SSc-ILD), Active systemic Juvenile idiopathic arthritis, Severe, active Rheumatoid arthritis

Tocilizumab (DB06273): A Comprehensive Monograph on its Pharmacology, Clinical Utility, and Regulatory Landscape

Executive Summary

Tocilizumab is a cornerstone biologic therapy that has fundamentally altered the treatment landscape for a spectrum of inflammatory and autoimmune disorders. Identified by DrugBank ID DB06273 and CAS Number 375823-41-9, it is a recombinant humanized monoclonal antibody of the immunoglobulin IgG1 subclass.[1] Its primary mechanism of action is the targeted inhibition of the interleukin-6 receptor (IL-6R), thereby neutralizing the pleiotropic effects of the pro-inflammatory cytokine IL-6.[1] By binding to both membrane-bound and soluble forms of the IL-6R, Tocilizumab provides a comprehensive blockade of IL-6-mediated signaling, a key driver in the pathophysiology of numerous diseases.[2]

Initially developed for rheumatologic conditions, Tocilizumab's clinical utility has expanded dramatically since its first approvals. It holds regulatory authorization in major markets for moderately to severely active rheumatoid arthritis (RA), giant cell arteritis (GCA), polyarticular juvenile idiopathic arthritis (pJIA), and systemic juvenile idiopathic arthritis (sJIA).[5] Its therapeutic reach extends beyond chronic autoimmune diseases to acute, life-threatening hyperinflammatory states, with landmark approvals for the management of chimeric antigen receptor (CAR) T-cell-induced cytokine release syndrome (CRS) and, more recently, for severe COVID-19 in hospitalized patients.[1] It is also approved for slowing pulmonary function decline in systemic sclerosis-associated interstitial lung disease (SSc-ILD).[5]

The pharmacologic profile of Tocilizumab is characterized by dose- and concentration-dependent pharmacokinetics, with a long terminal half-life that supports dosing intervals of two to four weeks.[9] A critical pharmacodynamic consideration is its indirect effect on the cytochrome P450 system; by suppressing inflammation, it can restore CYP enzyme activity, thereby increasing the metabolism and potentially reducing the efficacy of numerous co-administered drugs.[1]

The safety profile of Tocilizumab is intrinsically linked to its potent immunosuppressive mechanism. The drug carries a U.S. Food and Drug Administration (FDA) boxed warning—its most serious alert—for the risk of severe and potentially fatal infections, including tuberculosis, invasive fungal infections, and other opportunistic pathogens.[10] Other significant risks include gastrointestinal perforation, hepatotoxicity, neutropenia, and dyslipidemia, all of which necessitate careful patient selection and vigilant monitoring.[10]

Marketed principally as Actemra in the United States and RoActemra in Europe, Tocilizumab's commercial lifecycle has entered a new phase with the advent of approved biosimilars, such as Tofidence and Tyenne.[13] This development is poised to increase market competition and enhance patient access. Concurrently, extensive ongoing research continues to explore its potential in new therapeutic areas, most notably as an adjunctive therapy in oncology, underscoring its enduring relevance and evolving role in modern medicine.[16]

Molecular Profile and Mechanism of Action

Identity and Physicochemical Properties

Tocilizumab (DrugBank ID: DB06273; CAS Number: 375823-41-9) is a sophisticated biotech therapeutic classified as a recombinant humanized monoclonal antibody.[1] It belongs to the immunoglobulin G1 (IgG1) subclass and has been engineered to minimize immunogenicity while retaining high-affinity binding to its target.[2] The drug is known by several synonyms and development codes, including MRA (monoclonal antibody for RA), atlizumab, and R 1569.[17]

Commercially, it is primarily marketed under the brand name Actemra in the United States and RoActemra in the European Union and other international markets.[15] The maturation of its product lifecycle is underscored by the recent regulatory approval of several biosimilar versions, which are highly similar but not identical copies of the originator biologic. These include Tofidence (tocilizumab-bavi), Tyenne (tocilizumab-aazg), and Avtozma, signaling the end of market exclusivity and the beginning of a competitive landscape.[13]

Structurally, Tocilizumab is a large glycoprotein produced in Chinese Hamster Ovary (CHO) cells.[23] Its architecture consists of a human IgG1 constant framework onto which the complementarity-determining regions (CDRs)—the parts of the antibody that confer target specificity—from a murine (mouse) anti-human IL-6R antibody have been grafted.[2] This humanization process is critical for reducing the risk of patients developing an immune response against the drug itself. The complete molecule has a chemical formula of

C6428​H9976​N1720​O2018​S42​ and an approximate molar mass between 145 kDa and 148 kDa, with slight variations attributable to the natural heterogeneity of glycosylation, a post-translational modification common to antibodies.[1] The specific amino acid sequences for both the light and heavy chains of the antibody are well-documented and define its unique structure and function.[9]

The Interleukin-6 Pathway in Pathophysiology

To comprehend the therapeutic action of Tocilizumab, one must first understand the central role of its target, Interleukin-6 (IL-6). IL-6 is a pleiotropic cytokine, a type of signaling protein that has multiple, diverse effects on different cell types. It is produced by a wide array of cells, including T-cells, B-cells, monocytes, macrophages, and fibroblasts, typically in response to inflammatory stimuli such as infection or tissue injury.[3]

In a healthy state, IL-6 is a crucial mediator of the body's normal immune and inflammatory responses. It plays key roles in host defense, the regulation of hematopoiesis (blood cell formation), and the induction of the acute-phase response, a rapid systemic reaction to inflammation.[2] However, in many pathological states, the production of IL-6 becomes dysregulated and chronically elevated. This overproduction is a key pathogenic driver in a multitude of diseases, including autoimmune conditions like rheumatoid arthritis, systemic inflammatory syndromes such as Castleman's disease, and even certain malignancies.[1]

In the context of rheumatoid arthritis, for instance, IL-6 is found in high concentrations within the inflamed synovial tissue of the joints. There, it orchestrates a cascade of damaging effects: it promotes the differentiation of B-cells into autoantibody-producing plasma cells, enhances the infiltration of inflammatory cells into the joint, and, in synergy with other cytokines like TNF-α and IL-1, stimulates angiogenesis (the formation of new blood vessels) through increased production of vascular endothelial growth factor (VEGF).[2] Furthermore, IL-6 is instrumental in tipping the balance of T-helper cell differentiation towards the pro-inflammatory Th17 lineage, which is critically involved in the pathogenesis of arthritis.[2]

Systemically, the effects of excess IL-6 are profound. It acts on the liver to stimulate the production of acute-phase reactant proteins, such as C-reactive protein (CRP) and serum amyloid A (SAA), which are hallmark laboratory markers of systemic inflammation.[2] Simultaneously, it suppresses the production of other proteins like albumin and transferrin. This cascade of effects contributes to many of the constitutional symptoms of chronic inflammatory diseases, including fever, fatigue, anemia, and thrombocytosis (elevated platelet count).[2]

Molecular Mechanism of IL-6 Receptor Blockade

The biological activities of IL-6 are mediated through a unique receptor system that Tocilizumab is specifically designed to inhibit. The IL-6 receptor (IL-6R) exists in two principal forms: a membrane-bound receptor (mIL-6R) found on the surface of specific cells like hepatocytes and certain leukocytes, and a soluble form (sIL-6R) that circulates freely in the blood and extracellular fluid.[2]

Signaling is initiated when IL-6 binds to either mIL-6R or sIL-6R. This IL-6/IL-6R complex then associates with a second, separate membrane protein called glycoprotein 130 (gp130). The gp130 protein is the actual signal transducer and is expressed almost ubiquitously on cells throughout the body. The binding of the IL-6/IL-6R complex induces the homodimerization of two gp130 molecules, which in turn activates intracellular signaling cascades, most notably the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway.[2]

The therapeutic breadth of Tocilizumab can be attributed to its fundamental mechanism: the comprehensive inhibition of both of these signaling pathways. The signaling mediated by the membrane-bound receptor is often referred to as "classic signaling," while the pathway involving the soluble receptor is known as "trans-signaling".[4] Trans-signaling dramatically expands the range of IL-6's influence, allowing it to stimulate cells that do not express mIL-6R themselves but do have gp130 on their surface. Tocilizumab is a competitive antagonist that binds with high affinity to

both the soluble and membrane-bound forms of the IL-6R.[1] By occupying the receptor, it physically prevents IL-6 from binding and forming the initial complex. This blockade is the critical step that prevents the subsequent association with and dimerization of gp130, thereby completely shutting down the downstream JAK/STAT signaling cascade and halting the pro-inflammatory effects of IL-6.[1] The drug is so effective that it can even dissociate IL-6/IL-6R complexes that have already formed, ensuring a highly potent and thorough blockade of IL-6 signaling.[2] This dual blockade provides a more complete suppression of systemic inflammation than would be achieved by targeting only one pathway, explaining its efficacy in diseases characterized by widespread, multi-organ inflammation such as rheumatoid arthritis, sJIA, and CRS.

The arrival of biosimilars represents a pivotal moment in the history of Tocilizumab. Having first been approved in major Western markets around 2009-2010, the period of market exclusivity for the originator product, Actemra/RoActemra, has concluded.[5] The subsequent approval of biosimilars like Tofidence in 2023 and Tyenne in 2024 by regulatory bodies such as the FDA and EMA is predicated on a "totality of evidence" approach.[13] This rigorous process requires the biosimilar manufacturer to demonstrate high similarity in structure, purity, biological activity, and clinical pharmacokinetics, with no clinically meaningful differences in safety and efficacy compared to the reference product.[13] The introduction of these agents fosters a competitive market, which is anticipated to drive down costs, thereby increasing patient access and alleviating the substantial financial burden that high-cost biologic therapies place on healthcare systems globally.[14] It is important to note, however, that biosimilar approvals are not always a complete mirror of the originator; for instance, some biosimilars may not be approved for the full range of indications held by Actemra/RoActemra.[13]

Clinical Pharmacology

Pharmacodynamics

The pharmacodynamic effects of Tocilizumab are the direct physiological and biochemical consequences of its mechanism of action. The most immediate and measurable effect is a rapid and profound reduction in the levels of systemic inflammatory biomarkers. Treatment consistently leads to a marked decrease in the concentrations of acute-phase reactants, including C-reactive protein (CRP) and serum amyloid A (SAA), often normalizing them within weeks of initiating therapy.[2] This rapid normalization of CRP is a key indicator of therapeutic response and is used clinically to monitor drug activity. The drug also reverses other IL-6-driven systemic phenomena, such as thrombocytosis, anemia, and hypoalbuminemia, contributing to the improvement in patients' constitutional symptoms.[2]

A crucial and distinct pharmacodynamic property of Tocilizumab is its indirect interaction with the cytochrome P450 (CYP) enzyme system. Chronic inflammation, mediated by persistently high levels of IL-6, is known to suppress the expression and metabolic activity of various CYP isoenzymes.[1] By blocking IL-6 and quelling systemic inflammation, Tocilizumab reverses this suppression, leading to a normalization or effective up-regulation of CYP enzyme activity relative to the inflamed state. This is not a direct interaction with the enzymes themselves but an indirect consequence of its primary anti-inflammatory action. The clinical ramification is significant: the metabolism of co-administered drugs that are substrates for CYP enzymes (e.g., CYP3A4, CYP2C9, CYP1A2) can be increased, leading to lower serum concentrations and potentially reduced therapeutic efficacy.[1] This interaction has been observed with drugs such as simvastatin and oral contraceptives, and necessitates careful monitoring and potential dose adjustments of concomitant medications when Tocilizumab therapy is initiated or discontinued.[9]

Tocilizumab treatment is also associated with predictable changes in hematological parameters. Dose-related decreases in neutrophil counts (neutropenia) and platelet counts (thrombocytopenia) are well-documented adverse effects.[11] The precise mechanism underlying the neutropenia is not fully elucidated but is a recognized class effect of IL-6 pathway inhibition, given the role of IL-6 in hematopoiesis.[32] Furthermore, treatment frequently leads to elevations in lipid parameters, including total cholesterol, triglycerides, low-density lipoprotein (LDL), and high-density lipoprotein (HDL) cholesterol.[12] This effect necessitates routine monitoring of the patient's lipid profile during therapy.

Pharmacokinetics

The pharmacokinetic (PK) profile of Tocilizumab describes its absorption, distribution, metabolism, and elimination (ADME) within the body. As a large protein therapeutic, its PK properties differ substantially from those of small-molecule drugs.

Absorption: Following subcutaneous (SC) injection, Tocilizumab is absorbed slowly into the systemic circulation. The absorption half-life is estimated to be approximately 4 days in adult patients with RA and GCA, and 2 days in pediatric patients with JIA.[9] The peak serum concentration (

Cmax​) and total drug exposure (Area Under the Curve, AUC) are dependent on the dose and the frequency of administration.[9]

Distribution: Consistent with a large monoclonal antibody, Tocilizumab has a relatively small volume of distribution, indicating that it is primarily confined to the vascular and interstitial fluid compartments and does not extensively penetrate tissues. The pharmacokinetic profile, including the volume of distribution, varies meaningfully across different patient populations, which provides a clear rationale for the distinct, indication-specific dosing regimens. The volume of distribution at steady state (Vss​) has been characterized in several key populations: approximately 6.4 L in adults with RA, 7.46 L in adults with GCA, 4.08 L in pediatric patients with pJIA, and 4.01 L in pediatric patients with sJIA.[9] These differences in how the drug distributes throughout the body, coupled with variations in clearance, necessitate the tailored, often weight-based, dosing strategies to achieve comparable therapeutic exposures and clinical outcomes across these diverse patient groups. This demonstrates a sophisticated application of clinical pharmacology principles in establishing the drug's approved labeling.

Metabolism and Elimination: Tocilizumab is a protein and is therefore not metabolized by hepatic CYP enzymes. Instead, it is presumed to be broken down into smaller peptides and individual amino acids through general protein catabolism by proteolytic enzymes distributed throughout the body, in the same manner as endogenous IgG.[9]

The elimination of Tocilizumab is complex and exhibits concentration-dependency. At lower serum concentrations, the drug is primarily cleared via a saturable, target-mediated pathway (i.e., binding to IL-6R). As concentrations increase and the receptors become saturated, a non-saturable, linear proteolytic clearance pathway predominates. This results in dose-dependent clearance, which is non-linear at low doses and becomes linear at higher, therapeutic doses.[9] The terminal elimination half-life is also concentration-dependent, increasing as serum levels rise. In patients with RA, the effective half-life at steady state is approximately 21.5 days, which supports the every 2- to 4-week dosing intervals.[9] Total clearance is also population-dependent, with linear clearance values estimated at 12.5 mL/h in RA patients, 6.7 mL/h in GCA patients, 5.8 mL/h in pJIA patients, and 5.7 mL/h in sJIA patients.[9]

Table 3.2.1: Comparative Pharmacokinetic Parameters of Tocilizumab Across Key Patient Populations

ParameterRheumatoid Arthritis (Adult)Giant Cell Arteritis (Adult)Polyarticular JIA (Pediatric)Systemic JIA (Pediatric)Source(s)
Central Volume of Distribution (Vc​)3.5 L4.09 L1.98 L1.87 L9
Peripheral Volume of Distribution (Vp​)2.9 L3.37 L2.1 L2.14 L9
Volume of Distribution at Steady State (Vss​)6.4 L7.46 L4.08 L4.01 L9
Linear Clearance12.5 mL/h6.7 mL/h5.8 mL/h5.7 mL/h9
Absorption Half-life (SC)4 days4 days2 days2 days9
Terminal Half-life~21.5 days (concentration-dependent)8–14 days (concentration-dependent)Not specifiedNot specified1

Clinical Efficacy in Approved Indications

The clinical development of Tocilizumab showcases a strategic expansion from niche orphan diseases to broad, high-prevalence conditions, and ultimately to acute, life-threatening syndromes. This trajectory demonstrates the versatility of targeting a central cytokine like IL-6 and the capacity for rapid clinical development in response to urgent, unmet medical needs.

Rheumatoid Arthritis (RA)

Tocilizumab is indicated for the treatment of adult patients with moderately to severely active RA who have had an inadequate response to one or more disease-modifying antirheumatic drugs (DMARDs).[1] It can be administered as a monotherapy or in combination with conventional synthetic DMARDs such as methotrexate (MTX).[1]

The approval for RA was supported by a comprehensive global clinical development program that included five pivotal Phase III studies enrolling over 4,000 patients, with further studies supporting the subcutaneous formulation.[35] The evidence base is extensive, with numerous trials confirming its efficacy in various clinical scenarios. Studies such as NCT01258712 and NCT01235507 (ALABASTER) established its benefit when used in combination with MTX.[36] Other trials demonstrated its effectiveness in patients who had an inadequate response to either conventional DMARDs or more advanced TNF-alpha blockers (e.g., NCT00810277, NCT00144534), positioning it as a key option for difficult-to-treat RA.[36]

The evidence for Tocilizumab in RA has evolved significantly, challenging the traditional treatment hierarchy. Initially positioned as a later-line therapy, subsequent landmark trials elevated its status. The AMBITION study, for example, was a head-to-head comparison that demonstrated the statistical superiority of Tocilizumab monotherapy over MTX monotherapy—the long-standing cornerstone of RA treatment.[15] The LITHE study provided critical evidence that Tocilizumab could significantly reduce the rate of progression of structural joint damage as assessed by radiography.[15] More recently, a large-scale randomized clinical trial in China (NCT03155347) reaffirmed the superior efficacy of subcutaneous Tocilizumab, both as a monotherapy and in combination with MTX, compared to MTX alone.[37] This collective body of evidence has established Tocilizumab not merely as an alternative after TNF inhibitor failure, but as a viable first-line biologic, particularly for patients with contraindications or intolerance to MTX, or in whom monotherapy is preferred. The European label reflects this by including an indication for severe, active, and progressive RA in patients

not previously treated with MTX.[38]

Giant Cell Arteritis (GCA)

Tocilizumab is indicated for the treatment of GCA in adult patients, an approval that was a watershed moment in the management of this condition.[1] For over 50 years, high-dose corticosteroids were the only effective treatment, but their long-term use is associated with severe toxicity. Tocilizumab became the first therapy specifically approved by the FDA for GCA.[35]

The pivotal evidence for this indication came from the Phase III GiACTA study (NCT01791153), a global, randomized, double-blind, placebo-controlled trial involving 251 patients.[35] The trial was designed to assess the efficacy of Tocilizumab as a steroid-sparing agent. The results were compelling: at 52 weeks, a significantly higher proportion of patients receiving Tocilizumab (either weekly or every other week) in combination with a standardized 6-month prednisone taper achieved sustained remission compared to patients receiving placebo with a 26-week prednisone taper (56% and 53.1% vs. 14%, respectively).[35] This demonstrated a potent glucocorticoid-sparing effect, enabling patients to significantly reduce their cumulative steroid exposure while maintaining disease control, a primary goal of GCA therapy.[41]

Systemic & Polyarticular Juvenile Idiopathic Arthritis (sJIA & pJIA)

Tocilizumab is a key therapeutic option for two severe forms of childhood arthritis. It is approved for the treatment of active polyarticular juvenile idiopathic arthritis (pJIA) and active systemic juvenile idiopathic arthritis (sJIA) in patients aged two years and older.[1] For sJIA, it is specifically indicated for patients who have had an inadequate response to nonsteroidal anti-inflammatory drugs (NSAIDs) and systemic corticosteroids.[38] In both conditions, it can be used as a monotherapy or in combination with MTX.[1] The approvals were based on dedicated clinical trials establishing its efficacy and safety in this vulnerable pediatric population, with long-term data supporting its continued use (e.g., NCT00144625 for pJIA).[1]

Cytokine Release Syndrome (CRS)

In a significant expansion beyond autoimmune disease, Tocilizumab is approved for the treatment of chimeric antigen receptor (CAR) T-cell-induced severe or life-threatening CRS in both adults and pediatric patients two years of age and older.[1] CRS is a potentially fatal systemic inflammatory response triggered by the massive activation of CAR-T cells. IL-6 is a central mediator of this "cytokine storm."

The FDA approval was granted based on a retrospective analysis of pooled data from patients who developed CRS in the pivotal clinical trials of two pioneering CAR-T cell therapies, tisagenlecleucel (CTL019) and axicabtagene ciloleucel (KTE-C19).[7] In the primary efficacy cohort of 45 patients treated with tisagenlecleucel, 69% achieved a rapid and durable response, defined as resolution of CRS within 14 days of the first dose of Tocilizumab, with no more than two doses required.[7] This approval established Tocilizumab as the standard-of-care for managing a critical toxicity of a revolutionary class of cancer immunotherapies.

Systemic Sclerosis-Associated Interstitial Lung Disease (SSc-ILD)

Further demonstrating its utility in complex diseases with inflammatory and fibrotic components, Tocilizumab is indicated for slowing the rate of decline in pulmonary function in adult patients with SSc-ILD.[1] This is a rare and debilitating condition where fibrosis of the lungs leads to progressive respiratory failure. The approval was based on the results of the phase 3 focuSSced trial (NCT02453256), which built upon encouraging findings from the earlier phase 2 faSScinate study.[43] The trial demonstrated that while the primary endpoint of improvement in skin thickening was not met, Tocilizumab-treated patients showed a smaller decline in forced vital capacity (FVC), a key measure of lung function, compared to placebo, supporting its role in preserving lung health in this patient population.

COVID-19

Tocilizumab's role in the COVID-19 pandemic evolved from investigational use to a fully approved therapy. It is indicated for the treatment of hospitalized adult patients with COVID-19 who are receiving systemic corticosteroids and require supplemental oxygen, non-invasive or invasive mechanical ventilation, or extracorporeal membrane oxygenation (ECMO).[5]

The regulatory pathway involved an initial Emergency Use Authorization (EUA) from the FDA in June 2021, which was converted to a full approval in December 2022.[1] The European Medicines Agency (EMA) granted its approval in December 2021.[8] The approvals were based on the totality of evidence from four large randomized controlled trials involving over 5,500 hospitalized patients.[44] A key study was the REMAP-CAP trial, which showed that treatment with an IL-6 receptor antagonist (predominantly Tocilizumab) improved survival and reduced the time spent in intensive care for critically ill patients.[26] The main study evaluated by the EMA, involving 4,116 adults, found that Tocilizumab added to standard treatment (including corticosteroids) reduced the risk of death by 28 days from 35% in the standard care group to 31% in the Tocilizumab group.[8]

Table 4.1: Summary of Pivotal Clinical Trials for Tocilizumab by Indication

IndicationTrial Name/IdentifierPhaseDesignPatient PopulationPrimary EndpointKey Efficacy OutcomeSource(s)
Rheumatoid Arthritis (RA)AMBITIONIIIRCT, Double-BlindModerate-severe RA, MTX-intolerant/inappropriateACR20 response at Week 24TCZ monotherapy (70%) superior to MTX monotherapy (52%)15
Rheumatoid Arthritis (RA)LITHEIIIRCT, Double-BlindRA <5 yrs, inadequate MTX responseChange in Genant-modified Sharp score at Week 52TCZ + MTX significantly reduced radiographic progression vs. placebo + MTX15
Giant Cell Arteritis (GCA)GiACTA (NCT01791153)IIIRCT, Double-Blind, Placebo-ControlledAdult patients with GCAProportion of patients in sustained remission at Week 52TCZ + 6-month steroid taper (56%) superior to placebo + 26-week steroid taper (14%)35
Cytokine Release Syndrome (CRS)Retrospective Analysis (CTL019 trials)N/ARetrospective analysis of prospective trial dataPediatric & adult patients with CAR-T cell-induced severe/life-threatening CRSProportion of patients with CRS resolution within 14 days69% of patients achieved a response after 1-2 doses of TCZ7
Systemic Sclerosis-ILD (SSc-ILD)focuSSced (NCT02453256)IIIRCT, Double-Blind, Placebo-ControlledAdults with progressive SScChange in modified Rodnan Skin Score (mRSS)Primary endpoint not met, but TCZ showed less decline in FVC vs. placebo43
COVID-19REMAP-CAPIIIAdaptive Platform RCT, Open-LabelCritically ill, hospitalized adults with COVID-19Organ support-free daysIL-6 receptor antagonists improved survival and reduced organ support duration26
COVID-19RECOVERYIIIRCT, Open-LabelHospitalized adults with COVID-19 and hypoxia/inflammation28-day mortalityTCZ reduced mortality (29% vs 33%) and increased likelihood of discharge within 28 days8

Dosing, Administration, and Patient Management

The clinical application of Tocilizumab requires precise adherence to complex, indication-specific dosing regimens. This complexity underscores the narrow therapeutic window of the drug, where achieving a balance between efficacy and safety is paramount. The dosing strategies vary significantly by indication, patient age, body weight, and route of administration, reflecting the distinct pathophysiology and pharmacokinetic needs of each patient population.

Formulations and Routes of Administration

Tocilizumab is commercially available in two primary formulations to accommodate different clinical settings and patient preferences [12]:

  1. Intravenous (IV) Infusion: Supplied as a concentrate for solution, this formulation is administered as a 60-minute infusion in a clinical setting. It is the required route for acute conditions like CRS and COVID-19, and for the initial treatment of many of the chronic indications.[6]
  2. Subcutaneous (SC) Injection: This formulation is provided in a prefilled syringe or a single-dose prefilled autoinjector (e.g., ACTpen), designed for self-administration by the patient or a caregiver.[5] The SC route offers greater convenience and is an option for the chronic management of RA, GCA, JIA, and SSc-ILD, but it is not approved for all indications or patient populations.[48]

Recommended Dosing Regimens

The recommended dosing for Tocilizumab is highly specific and is detailed in the table below. These regimens have been established through extensive clinical trials to optimize the risk-benefit profile for each condition.

Table 5.1: Recommended Dosing and Administration of Tocilizumab for Approved U.S. Indications

IndicationPatient PopulationRouteRecommended DoseFrequencyMaximum Dose / NotesSource(s)
Rheumatoid Arthritis (RA)AdultsIV4 mg/kg, may increase to 8 mg/kgEvery 4 weeksDo not exceed 800 mg per infusion.11
Adults <100 kgSC162 mgEvery other week, may increase to every week34
Adults ≥100 kgSC162 mgEvery week34
Giant Cell Arteritis (GCA)AdultsIV6 mg/kgEvery 4 weeksDo not exceed 600 mg per infusion. Used with tapering glucocorticoids.48
AdultsSC162 mgEvery week or every other weekBased on clinical considerations. Used with tapering glucocorticoids.48
Polyarticular JIA (pJIA)Pediatric (<30 kg)IV10 mg/kgEvery 4 weeks39
Pediatric (≥30 kg)IV8 mg/kgEvery 4 weeks39
Pediatric (<30 kg)SC162 mgEvery 3 weeks48
Pediatric (≥30 kg)SC162 mgEvery 2 weeks48
Systemic JIA (sJIA)Pediatric (<30 kg)IV12 mg/kgEvery 2 weeks11
Pediatric (≥30 kg)IV8 mg/kgEvery 2 weeks11
Pediatric (<30 kg)SC162 mgEvery 2 weeks48
Pediatric (≥30 kg)SC162 mgEvery week48
Cytokine Release Syndrome (CRS)All patients (<30 kg)IV12 mg/kgSingle doseMay repeat up to 3 times if needed, at least 8 hours apart.7
All patients (≥30 kg)IV8 mg/kgSingle doseDo not exceed 800 mg per dose.7
Systemic Sclerosis-ILD (SSc-ILD)AdultsSC162 mgEvery week5
COVID-19Hospitalized AdultsIV8 mg/kgSingle 60-min infusionDo not exceed 800 mg.6

Dose Modifications and Patient Monitoring

Safe use of Tocilizumab requires proactive patient management, including baseline screening and ongoing monitoring for potential toxicities.

Initiation Criteria: Treatment should not be initiated in patients with certain laboratory abnormalities. Key contraindications to starting therapy include an absolute neutrophil count (ANC) below 2000 per mm3, a platelet count below 100,000 per mm3, or alanine aminotransferase (ALT) or aspartate aminotransferase (AST) levels above 1.5 times the upper limit of normal (ULN).[11]

Routine Monitoring: Due to the known risks of hematologic and hepatic adverse events, regular laboratory monitoring is mandatory. This includes a complete blood count with differential (to monitor neutrophils and platelets) and a liver function panel.[12] A lipid panel should also be checked 4 to 8 weeks after initiating therapy and periodically thereafter.

Dose Adjustments for Abnormalities: The prescribing information provides explicit guidance for managing treatment-related laboratory changes. For example, in RA patients, the development of severe neutropenia (ANC < 1000) or thrombocytopenia (< 50,000), or significant elevations in liver enzymes (ALT/AST > 3x ULN) would necessitate dose interruption. For less severe abnormalities, a dose reduction may be recommended, such as decreasing the IV dose from 8 mg/kg to 4 mg/kg or increasing the dosing interval of the SC formulation.[11]

Safety Profile and Risk Mitigation

The safety profile of Tocilizumab is a direct and logical extension of its potent mechanism of action. The adverse events are not random occurrences but are predictable consequences of blocking the IL-6 pathway, a central signaling axis in the immune system. This direct link between mechanism and risk means that adverse events are an inherent trade-off for therapeutic benefit, making risk mitigation through careful screening, monitoring, and patient education a cornerstone of its clinical use.

Boxed Warning: Serious Infections

Tocilizumab carries a boxed warning, the most stringent warning issued by the U.S. FDA, regarding the increased risk of serious infections that may lead to hospitalization or death.[10] By inhibiting IL-6, the drug compromises the body's ability to mount an effective defense against a wide range of pathogens.

Types of Infections: The reported infections are diverse and include:

  • Tuberculosis (TB): Both new-onset active TB and reactivation of latent TB have been observed, which can present as either pulmonary or extrapulmonary disease.[10]
  • Invasive Fungal Infections: Life-threatening infections such as candidiasis, aspergillosis, and pneumocystis have occurred. A key clinical feature is that these infections may present with disseminated rather than localized disease, making diagnosis challenging.[10]
  • Bacterial and Viral Infections: Common bacterial infections (e.g., cellulitis, pneumonia) and viral infections (e.g., herpes zoster) occur at an increased frequency.[33]
  • Opportunistic Infections: Infections caused by pathogens that do not typically cause disease in immunocompetent individuals are a significant risk.[10]

Risk Mitigation Strategies:

  • Screening: Before initiating Tocilizumab (except in the acute setting of COVID-19), all patients must be screened for latent TB infection. If the test is positive, treatment for latent TB should be started before commencing Tocilizumab.[10] Screening for viral hepatitis (particularly hepatitis B) is also critical, as HBV reactivation has been reported and can lead to fulminant hepatitis.[29]
  • Vigilant Monitoring: Patients must be closely monitored for any signs or symptoms of infection (e.g., fever, cough, fatigue, skin sores) both during and after treatment. It is crucial to consider the possibility of TB even in patients who initially tested negative for latent infection.[10] If a serious infection develops, Tocilizumab must be interrupted until the infection is controlled.[10]

Other Significant Adverse Reactions

Beyond the risk of infection, several other serious adverse events are associated with Tocilizumab therapy.

  • Gastrointestinal (GI) Perforation: Events of lower intestinal perforation have been reported, primarily as a complication of diverticulitis in RA patients.[1] The mechanism is thought to involve the impairment of mucosal healing and containment of localized inflammation. The drug should be used with caution in patients with a history of diverticulitis, and any patient presenting with new-onset abdominal pain, fever, and changes in bowel habits must be promptly evaluated for perforation.[6]
  • Hepatotoxicity: Serious cases of drug-induced liver injury, some resulting in acute liver failure requiring transplant or leading to death, have been observed with both IV and SC formulations.[10] The onset can be insidious, occurring months to years after treatment initiation. Regular monitoring of liver function tests is essential, and the drug should be interrupted if clinically significant abnormalities develop.[10]
  • Hematological Abnormalities: As noted previously, dose-related neutropenia and thrombocytopenia are known risks that require routine blood count monitoring.[11]
  • Dyslipidemia: Clinically significant increases in lipid parameters, including total cholesterol, LDL, HDL, and triglycerides, are common and may require initiation or adjustment of lipid-lowering therapy.[12]
  • Hypersensitivity Reactions: Serious hypersensitivity reactions, including anaphylaxis, have been reported. Fatal anaphylactic events have occurred with the IV infusion. Therefore, IV Tocilizumab should only be administered by healthcare professionals with appropriate medical support available. The drug is strictly contraindicated in patients with a known prior hypersensitivity reaction.[10]
  • Malignancy Risk: As with other potent immunosuppressants that alter immune surveillance, there is a theoretical risk that Tocilizumab may increase the incidence of certain malignancies. This risk remains a subject of long-term observation.[29]
  • Demyelinating Disorders: Rare cases of new-onset or exacerbation of central nervous system demyelinating disorders, such as multiple sclerosis, have been reported.[29]

A notable point of discussion has been the potential for unlisted cardiovascular risks. While competing TNF-blocker therapies carry label warnings for side effects like heart failure, the Actemra label historically did not, which may have led to a perception of a more favorable cardiovascular safety profile.[29] However, post-marketing surveillance and reports to the FDA have raised concerns about potential risks of heart failure, heart attack, and stroke in patients taking Tocilizumab.[29] This discrepancy highlights a common challenge in pharmacovigilance, where rare or long-term adverse events may only become apparent after widespread use in a real-world population, beyond the confines of pre-approval clinical trials.

Contraindications and Precautions

  • Contraindication: The only absolute contraindication is known hypersensitivity to Tocilizumab or any of its excipients.[10]
  • Vaccinations: Patients should not receive live or live-attenuated vaccines concurrently with Tocilizumab. It is recommended that all vaccinations be brought up to date according to current immunization guidelines prior to starting therapy.[11]
  • Combination with Other Biologics: The concomitant use of Tocilizumab with other biologic DMARDs (e.g., TNF antagonists, abatacept, rituximab) or JAK inhibitors is not recommended, as this could lead to additive immunosuppression and an unacceptably high risk of serious infection.[11]

Drug-Drug Interactions

Tocilizumab has a unique and extensive drug-drug interaction profile, primarily driven by its pharmacodynamic effect on CYP450 enzymes.[9]

  • CYP450 Substrates: By reversing the inflammation-induced suppression of CYP enzymes, Tocilizumab can increase the metabolism of a vast number of drugs that are CYP substrates. This can lead to decreased exposure and potentially reduced efficacy of these concomitant medications. This interaction affects a wide range of drug classes, including statins (simvastatin, atorvastatin), anticoagulants (warfarin), immunosuppressants (cyclosporine), cardiovascular drugs (diltiazem), and hormonal contraceptives. Close monitoring and potential dose increases of these agents may be necessary upon initiation of Tocilizumab.[1]
  • Additive Immunosuppression: Combining Tocilizumab with other immunosuppressive or immunomodulatory agents, including corticosteroids, methotrexate, and other biologics, increases the overall degree of immunosuppression and elevates the risk of infection and other adverse events.[9]
  • Vaccine Efficacy: Due to its effect on the immune system, Tocilizumab may reduce the immunogenicity and therapeutic efficacy of both live and inactivated vaccines.[9]

Regulatory and Commercial Landscape

Global Development and Regulatory History

The story of Tocilizumab is a prime example of successful translational medicine, originating from fundamental academic research and culminating in a globally significant therapeutic. The foundational science was laid at Osaka University in Japan, where Professor Tadamitsu Kishimoto and his team discovered and cloned IL-6 and its receptor in the 1980s.[1] This basic science discovery paved the way for a therapeutic application.

The drug itself was jointly developed by Osaka University and Chugai Pharmaceutical, a Japanese company that initiated clinical development for RA in 1997.[1] Chugai achieved the world's first regulatory approval for Tocilizumab in Japan in June 2005, for the treatment of Castleman's disease, a rare lymphoproliferative disorder.[1]

The global success of Tocilizumab was catalyzed by a pivotal co-development and licensing agreement between Chugai and the Swiss multinational healthcare company Hoffmann-La Roche in 2003.[1] This partnership was instrumental, combining Chugai's innovative science with Roche's vast global resources for large-scale clinical trials, regulatory navigation, and commercialization. Under the agreement, Chugai retains marketing rights in Japan and some other Asian nations, while Roche markets the drug in the rest of the world, including the major US and European markets.[1] This symbiotic relationship—Japanese innovation fused with global development power—was key to transforming a Japan-first drug into a global standard of care available in over 110 countries.[53]

Major Market Approvals:

  • European Union: The European Medicines Agency (EMA) granted marketing authorization for RoActemra in January 2009 for the treatment of RA.[1] The label has since been expanded numerous times, with a notable extension for severe COVID-19 granted in December 2021.[8]
  • United States: The U.S. Food and Drug Administration (FDA) approved Actemra in January 2010 for RA.[5] The path to approval included an initial Complete Response Letter from the FDA in 2008, which was subsequently addressed.[5]

The regulatory history of Tocilizumab in the U.S. is a case study in effective lifecycle management. Following the initial RA approval, the developers pursued a strategic series of supplemental applications for new indications, often for conditions with high unmet need, which allowed them to secure expedited review pathways like Breakthrough Therapy Designation and Priority Review.[35] This led to a cascade of approvals: sJIA (2011), pJIA (2013), GCA (2017), CRS (2017), SSc-ILD (2021), and full approval for COVID-19 (2022).[5] In parallel, new formulations were developed and approved, including a subcutaneous version (2013) and a patient-friendly autoinjector (2018), which enhanced convenience and solidified its market position.[5] This systematic, evidence-driven expansion transformed Tocilizumab from a niche product into a blockbuster drug with a broad therapeutic footprint.

The Emergence of Biosimilars

With the expiration of its core patents, the Tocilizumab market has entered a new era of competition with the introduction of biosimilars.

  • U.S. Market Entry: The first tocilizumab biosimilar to receive FDA approval was Biogen's Tofidence (tocilizumab-bavi) in September 2023.[14] This was followed by the approval of Fresenius Kabi's Tyenne (tocilizumab-aazg) in March 2024.[13]
  • European Market Entry: The EMA has also reviewed and approved biosimilars, including Tyenne and Avtozma, based on applications demonstrating their high similarity to the reference product, RoActemra.[21]
  • Regulatory Framework: The approval of a biosimilar is a rigorous scientific process. The manufacturer must submit a comprehensive data package that includes extensive analytical characterization to demonstrate structural and functional similarity, as well as non-clinical and clinical studies to confirm comparable pharmacokinetics, efficacy, safety, and immunogenicity to the originator product.[13]
  • Key Distinctions: It is important to note key regulatory nuances. A biosimilar is not automatically "interchangeable" with the reference product. An interchangeability designation, which Tyenne does not have, would allow a pharmacist to substitute it for Actemra without consulting the prescriber.[13] Furthermore, biosimilars may not be approved for the entire slate of indications held by the originator. For instance, at the time of its approval, Tyenne was not indicated for SSc-ILD, an indication for which Actemra is approved.[13]

Investigational Applications and Future Directions

The established efficacy of Tocilizumab in its approved indications has spurred extensive research into its potential use in a wide array of other diseases. This breadth of investigation highlights a paradigm shift in the perception of the drug: it is viewed less as a specific "anti-rheumatic" agent and more as a broad-spectrum "inflammation modulator" with potential utility across numerous medical disciplines.

Off-Label and Investigational Uses in Rheumatic and Autoimmune Diseases

Given the central role of IL-6 in many autoimmune and autoinflammatory processes, Tocilizumab has been studied or used off-label in a variety of conditions beyond its approved indications.

  • Systemic Lupus Erythematosus (SLE): Elevated IL-6 levels are correlated with disease activity in SLE. A small Phase I study and several case reports have suggested that Tocilizumab can improve arthritis and other inflammatory manifestations of SLE, although dose-dependent neutropenia was a notable safety concern.[56]
  • Other Conditions: The literature contains reports on the investigational use of Tocilizumab in a diverse range of diseases, including Takayasu arteritis, juvenile dermatomyositis, juvenile localized scleroderma, autoimmune encephalitis, refractory Kawasaki's disease, and JIA-associated uveitis.[15] It has also shown promise in rare hereditary autoinflammatory conditions like Tumor Necrosis Factor Receptor-Associated Periodic Syndrome (TRAPS) and H syndrome.[56]

Investigational Uses in Oncology

Perhaps the most dynamic area of current research for Tocilizumab is in oncology. Beyond its established supportive care role in managing CRS, it is now being actively investigated as a direct adjunctive anti-cancer therapy. The rationale is that IL-6 is deeply implicated in cancer biology, contributing to tumor cell proliferation, survival, angiogenesis, and the creation of an immunosuppressive tumor microenvironment that can blunt the efficacy of immunotherapies.

A review of active clinical trial registries reveals a robust pipeline of studies evaluating Tocilizumab in combination regimens for various cancers [16]:

  • Solid Tumors: Trials are underway combining Tocilizumab with chemotherapy (e.g., carboplatin for triple-negative breast cancer) or with checkpoint inhibitors (e.g., atezolizumab for prostate cancer, head and neck cancer, and non-small cell lung cancer).
  • Hematologic Malignancies: It is being studied in combination with other biologics like the bispecific antibody glofitamab for B-cell non-Hodgkin lymphoma, and with targeted therapies like the BCL-2 inhibitor venetoclax for multiple myeloma.
  • Pediatric Oncology: An active trial is investigating its use for progressive or recurrent pediatric adamantinomatous craniopharyngioma, a rare and difficult-to-treat brain tumor.[16]

This pattern of investigation suggests that the future of Tocilizumab may lie less in securing new monotherapy indications and more in its synergistic role within complex combination regimens. In fields like immuno-oncology, the strategy is not to replace existing standards of care but to enhance their efficacy or mitigate their toxicity. By modulating the inflammatory tumor microenvironment, Tocilizumab may overcome resistance to checkpoint inhibitors or, as in CRS, enable the safe use of other powerful therapies. This positions Tocilizumab as a key "enabling" therapy that could significantly extend its clinical utility long after the expiration of its primary patents.

Critical Analysis and Concluding Remarks

The COVID-19 Efficacy Controversy

While Tocilizumab ultimately gained approval for treating severe COVID-19, the journey to that approval was marked by significant controversy and conflicting clinical trial data. This situation highlighted the immense challenges of conducting rigorous clinical research during a rapidly evolving global pandemic.

On one hand, large, well-designed platform trials like REMAP-CAP and RECOVERY, as well as the main study reviewed by the EMA, demonstrated a modest but statistically significant mortality benefit in a specific population: hospitalized patients with severe COVID-19, hypoxia, and systemic inflammation who were also receiving corticosteroids.[8] However, other studies produced contradictory results. A randomized trial in Brazil was stopped early by its data safety board due to a signal of

increased mortality in the Tocilizumab arm (17% vs. 3%).[46] Other prominent trials, including COVACTA, EMPACTA, and REMDACTA, failed to meet their primary endpoints for improving clinical status or survival, although some showed benefits on secondary endpoints like reducing progression to mechanical ventilation.[46]

Several factors may explain these discrepancies, including differences in trial design, patient populations (e.g., severity of illness at enrollment), timing of drug administration (early vs. late in the disease course), and, critically, the concomitant use of other therapies, especially systemic corticosteroids, which later became the standard of care.[46] The small size of some trials, such as the one in Brazil, meant that the alarming signal of harm could have been due to statistical chance.[46] Ultimately, regulatory agencies based their approvals on the "totality of evidence," which, when synthesized in large meta-analyses, tilted in favor of a clear, albeit modest, benefit for the specific, narrowly defined patient population now reflected in the drug's label.[8]

Balancing Therapeutic Benefit and Immunosuppressive Risk

The central clinical challenge in utilizing Tocilizumab is navigating the inherent and inseparable balance between its profound therapeutic benefits and its significant immunosuppressive risks. The drug is undeniably effective in controlling debilitating, disabling, and sometimes life-threatening inflammatory diseases.[2] This efficacy, however, is achieved by potently suppressing a key component of the immune system, which comes at the direct cost of an increased risk of serious infections, GI perforation, and other major adverse events.[10]

This "double-edged sword" nature is particularly acute in the context of infectious diseases like COVID-19. While inhibiting the IL-6-driven "cytokine storm" can prevent catastrophic organ damage, the same inhibition may impair the immune system's ability to clear the primary viral pathogen or to fight off secondary bacterial or fungal superinfections that are common in critically ill patients.[32] This delicate balance necessitates extremely careful patient selection, vigilant monitoring for complications, and a high degree of clinical acumen to ensure that the potential benefits outweigh the substantial risks in any given patient.

Concluding Perspective

Tocilizumab stands as a landmark achievement in modern pharmacology and a testament to the power of translational medicine. Its journey from the fundamental scientific discovery of a cytokine and its receptor in a university laboratory to a globally impactful therapeutic agent has transformed the standard of care for millions of patients. The drug's development history exemplifies a masterful strategy of evidence-based indication expansion and intelligent lifecycle management.

Its potent and specific mechanism of action is the source of both its remarkable efficacy and its significant, predictable risk profile, demanding a sophisticated approach to patient management. As Tocilizumab enters a new phase of its lifecycle, the emergence of biosimilars is poised to democratize access to this vital therapy, while a vibrant and expanding program of investigational research continues to probe its potential in new therapeutic frontiers, particularly in the synergistic treatment of cancer. The story of Tocilizumab is a continuing narrative of scientific innovation, clinical evolution, and enduring therapeutic relevance.

Works cited

  1. Tocilizumab - Wikipedia, accessed July 10, 2025, https://en.wikipedia.org/wiki/Tocilizumab
  2. Review of tocilizumab in the treatment of rheumatoid arthritis - PMC, accessed July 10, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2727785/
  3. What is the mechanism of Tocilizumab? - Patsnap Synapse, accessed July 10, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-tocilizumab
  4. Tocilizumab | Anti-Human IL6R |COVID-19 - TargetMol, accessed July 10, 2025, https://www.targetmol.com/compound/tocilizumab
  5. Actemra (tocilizumab) FDA Approval History - Drugs.com, accessed July 10, 2025, https://www.drugs.com/history/actemra.html
  6. ACTEMRA® (tocilizumab) Treatment Information for Healthcare Professionals, accessed July 10, 2025, https://www.actemrahcp.com/
  7. FDA Approval Summary: Tocilizumab for Treatment of Chimeric Antigen Receptor T Cell‐Induced Severe or Life‐Threatening Cytokine Release Syndrome | The Oncologist | Oxford Academic, accessed July 10, 2025, https://academic.oup.com/oncolo/article-abstract/23/8/943/6439647
  8. European Commission Approves Indication Extension for Monoclonal Antibody RoActemra (Tocilizumab) for Treatment of Severe COVID-19 - Paul-Ehrlich-Institut, accessed July 10, 2025, https://www.pei.de/EN/newsroom/hp-news/2021/211209-indication-extension-roactemra-treatment-covid-19.html
  9. Tocilizumab: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed July 10, 2025, https://go.drugbank.com/drugs/DB06273
  10. Important Safety Information for Giant Cell Arteritis HCPs | ACTEMRA® (tocilizumab) for GCA, accessed July 10, 2025, https://www.actemrahcp.com/gca/safety.html
  11. Reference ID: 4077494 This label may not be the latest approved by FDA. For current labeling information, please visit https://, accessed July 10, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125276s111,125472s023lbl.pdf
  12. Tocilizumab (Actemra) - American College of Rheumatology, accessed July 10, 2025, https://rheumatology.org/patients/tocilizumab-actemra
  13. Tyenne (tocilizumab-aazg) FDA Approval History - Drugs.com, accessed July 10, 2025, https://www.drugs.com/history/tyenne.html
  14. FDA Approves Biogen's TOFIDENCE™ (tocilizumab-bavi), a Biosimilar Referencing ACTEMRA® | Nasdaq, accessed July 10, 2025, https://www.nasdaq.com/press-release/fda-approves-biogens-tofidencetm-tocilizumab-bavi-a-biosimilar-referencing-actemrar
  15. Tocilizumab (Actemra) - PMC - PubMed Central, accessed July 10, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5612212/
  16. Clinical Trials Using Tocilizumab - NCI, accessed July 10, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/tocilizumab?pn=1
  17. Tocilizumab (MRA; atlizumab) | CAS 375823-41-9 | AbMole BioScience, accessed July 10, 2025, https://www.abmole.com/products/tocilizumab.html
  18. Tocilizumab [USAN:INN] - PubChem, accessed July 10, 2025, https://pubchem.ncbi.nlm.nih.gov/substance/135345962
  19. Tocilizumab - Chugai Pharmaceutical/Roche - AdisInsight - Springer, accessed July 10, 2025, https://adisinsight.springer.com/drugs/800010359
  20. Definition of tocilizumab - NCI Drug Dictionary, accessed July 10, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/tocilizumab
  21. Avtozma | European Medicines Agency (EMA), accessed July 10, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/avtozma
  22. Tocilizumab - brand name list from Drugs.com, accessed July 10, 2025, https://www.drugs.com/ingredient/tocilizumab.html
  23. Tocilizumab (anti-IL-6R) IL Receptor inhibitor | Read Reviews & Product Use Citations, accessed July 10, 2025, https://www.selleckchem.com/products/tocilizumab.html
  24. Tocilizumab (Synonyms: Anti-Human IL6R, Humanized Antibody) - MedchemExpress.com, accessed July 10, 2025, https://www.medchemexpress.com/tocilizumab.html
  25. pmc.ncbi.nlm.nih.gov, accessed July 10, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2727785/#:~:text=Tocilizumab%20is%20a%20humanized%20anti,transmembrane%20signaling%20of%20IL%2D6.
  26. tocilizumab | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed July 10, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=refs&ligandId=6881
  27. Tocilizumab: A Novel Humanized Anti-Interleukin 6 Receptor Antibody for the Treatment of Patients with Rheumatoid Arthritis, accessed July 10, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3389389/
  28. Table 5, Key Limitations - Tocilizumab (Actemra) - NCBI Bookshelf, accessed July 10, 2025, https://www.ncbi.nlm.nih.gov/books/NBK349519/table/T47/
  29. Actemra | Uses, Safety Info & Comparison to Other RA Drugs - Drugwatch.com, accessed July 10, 2025, https://www.drugwatch.com/actemra/
  30. Tocilizumab Completed Phase 1 Trials for Rheumatoid Arthritis Treatment - DrugBank, accessed July 10, 2025, https://go.drugbank.com/drugs/DB06273/clinical_trials?conditions=DBCOND0027961&phase=1&purpose=treatment&status=completed
  31. How is Actemra® (tocilizumab) for GCA believed to work?, accessed July 10, 2025, https://www.actemra.com/gca/considerations/how-it-works.html
  32. The Controversies Surrounding Tocilizumab Administration Following Pathogen-Associated Molecular Pattern (PAMP) Induced by COVID-19 - KnE Publishing, accessed July 10, 2025, https://publish.kne-publishing.com/index.php/JPC/article/download/4549/4268/
  33. Tocilizumab | Side-effects, uses, time to work - Versus Arthritis, accessed July 10, 2025, https://versusarthritis.org/about-arthritis/treatments/drugs/tocilizumab/
  34. Actemra (tocilizumab) injection label - accessdata.fda.gov, accessed July 10, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125276s092lbl.pdf
  35. FDA Approves Genentech's Actemra (Tocilizumab) for Giant Cell Arteritis, accessed July 10, 2025, https://www.gene.com/media/press-releases/14667/2017-05-22/fda-approves-genentechs-actemra-tocilizu
  36. Rheumatoid Arthritis Completed Phase 3 Trials for Tocilizumab (DB06273) - DrugBank, accessed July 10, 2025, https://go.drugbank.com/indications/DBCOND0027961/clinical_trials/DB06273?phase=3&status=completed
  37. Tocilizumab Monotherapy or Combined With Methotrexate for Rheumatoid Arthritis: A Randomized Clinical Trial - PubMed, accessed July 10, 2025, https://pubmed.ncbi.nlm.nih.gov/40388166/
  38. RoActemra | European Medicines Agency (EMA), accessed July 10, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/roactemra
  39. ACTEMRA (tocilizumab) injection - accessdata.fda.gov, accessed July 10, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125276s114lbl.pdf
  40. Tocilizumab approved for giant cell arteritis - American Academy of Ophthalmology, accessed July 10, 2025, https://www.aao.org/education/headline/tocilizumab-approved-giant-cell-arteritis
  41. GCA Clinical Trial Results | ACTEMRA® (tocilizumab), accessed July 10, 2025, https://www.actemra.com/gca/considerations/clinical-trial-results.html
  42. Tocilizumab Completed Phase 3 Trials for polyarticular Juvenile Idiopathic Arthritis Treatment | DrugBank Online, accessed July 10, 2025, https://go.drugbank.com/drugs/DB06273/clinical_trials?conditions=DBCOND0025601&phase=3&purpose=treatment&status=completed
  43. A Review of Recent Advances Using Tocilizumab in the Treatment of Rheumatic Diseases, accessed July 10, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5935615/
  44. Actemra/RoActemra approved by the European Commission to treat patients with severe COVID-19 - Roche, accessed July 10, 2025, https://www.roche.com/investors/updates/inv-update-2021-12-07
  45. EMA recommends approval for use of RoActemra in adults with severe COVID-19, accessed July 10, 2025, https://www.ema.europa.eu/en/news/ema-recommends-approval-use-roactemra-adults-severe-covid-19
  46. Covid-19 controversies: the tocilizumab chapter - The BMJ, accessed July 10, 2025, https://www.bmj.com/content/372/bmj.n244
  47. tocilizumab (Actemra®) - Blue Shield of California, accessed July 10, 2025, https://www.blueshieldca.com/bin/cms/bsca/services/portal/provider/StreamDocumentServlet?fileName=PRV_tocilizumab-Actemra-COMM-Rx-Provider.pdf
  48. ACTEMRA® (tocilizumab) injection, for ... - accessdata.fda.gov, accessed July 10, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/125276s144,125472s056lbl.pdf
  49. Actemra® (tocilizumab) – Updated labeling, accessed July 10, 2025, https://professionals.optumrx.com/content/dam/optum3/professional-optumrx/news/rxnews/clinical-updates/clinicalupdate_actemra_2022-0303.pdf
  50. Update on tocilizumab in rheumatoid arthritis: a narrative review - Frontiers, accessed July 10, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2025.1470488/full
  51. Tocilizumab Uses, Side Effects & Warnings - Drugs.com, accessed July 10, 2025, https://www.drugs.com/mtm/tocilizumab.html
  52. Tocilizumab (Actemra®): Uses & Side Effects - Cleveland Clinic, accessed July 10, 2025, https://my.clevelandclinic.org/health/drugs/18763-tocilizumab-injection
  53. Antibody Drug Discovery - Continuous Innovative Drug Discovery | History of Chugai Pharmaceutical | Understanding Chugai: Our Company and History | About Chugai | CHUGAI PHARMACEUTICAL CO., LTD., accessed July 10, 2025, https://www.chugai-pharm.co.jp/english/profile/overview/history/history06.html
  54. Tocilizumab - PubMed, accessed July 10, 2025, https://pubmed.ncbi.nlm.nih.gov/20065633/
  55. EMA application for a tocilizumab biosimilar accepted - Fresenius Kabi, accessed July 10, 2025, https://www.fresenius-kabi.com/news/fresenius-kabi-tocilizumab-mma-accepted-by-ema
  56. Off-label use of tocilizumab to treat non-juvenile idiopathic arthritis in pediatric rheumatic patients: a literature review - PubMed Central, accessed July 10, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6295005/
  57. RoActemra (tocilizumab) approved by EMA for use in severe COVID-19, accessed July 10, 2025, https://hospitalhealthcare.com/covid-19/roactemra-approved-by-ema-for-use-in-severe-covid-19/

Published at: July 10, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.