PEPID Connect Help
View Tutorial
Contact PEPID Support
Suggest an edit
Current tool:
Current monograph:
Hello, PEPID User
PEPID
Subsections
Tocilizumab

TOC

Tocilizumab

Class: 90:24.20.92 Interleukin-mediated Agents, Miscellaneous

  Black Box Warning

    Serious Infections
  • Serious, sometimes fatal infections including tuberculosis (pulmonary or extrapulmonary disease), bacterial and viral infections, invasive fungal infections (may be disseminated), and other opportunistic infections reported.(See Infectious Complications under Cautions.)

  • Carefully consider risks and benefits prior to initiating tocilizumab therapy in patients with chronic or recurring infections.

  • Evaluate patients for latent tuberculosis infection prior to and periodically during tocilizumab therapy; if indicated, initiate appropriate antimycobacterial regimen prior to initiating tocilizumab therapy.

  • Closely monitor patients for infection, including active tuberculosis in those with a negative tuberculin skin test, during and after treatment. If serious infection develops, discontinue tocilizumab until infection is controlled.

Introduction

Tocilizumab, a recombinant humanized immunoglobulin G1 (IgG1) monoclonal antibody specific for the interleukin-6 (IL-6) receptor, is a biologic response modifier and a disease-modifying antirheumatic drug (DMARD).(1)(4)

Uses

Tocilizumab is used alone or in combination with methotrexate or other nonbiologic disease-modifying antirheumatic drugs (DMARDs) for the management of moderately to severely active rheumatoid arthritis in adults who have had an inadequate response to one or more DMARDs(1)(3)(4)(5)(6)(7)(29) and also is used alone or in combination with methotrexate for the management of active systemic or polyarticular juvenile idiopathic arthritis (JIA).(1)(20)(21) The drug is used in the management of giant cell arteritis (GCA) in adults.(1)(50)(51)(52)(53) Tocilizumab is also used in the management of chimeric antigen receptor (CAR) T cell-induced severe or life-threatening cytokine release syndrome (CRS).(1)(61) Tocilizumab is also used in adults with systemic sclerosis-associated interstitial lung disease (SSc-ILD) to slow the rate of decline in pulmonary function.(1)(80)(81) In addition, the drug is used in the management of coronavirus disease 2019 (COVID-19) in hospitalized patients.(1)(31)(32)(33)(41)(43)(44)(47)(48)

Rheumatoid Arthritis in Adults

Tocilizumab is used for the management of moderately to severely active rheumatoid arthritis in adults who have had an inadequate response to one or more DMARDs.(1) Tocilizumab can be used alone or in combination with methotrexate or other nonbiologic DMARDs (e.g., hydroxychloroquine, leflunomide, minocycline, sulfasalazine).(1)(3)(4)(5)(6)(7)(29) Concomitant use of tocilizumab with other biologic DMARDs, such as tumor necrosis factor (TNF; TNF-α) blocking agents (e.g., adalimumab, certolizumab pegol, etanercept, golimumab, infliximab), interleukin-1 (IL-1) receptor antagonists (e.g., anakinra), anti-CD20 monoclonal antibodies (e.g., rituximab), and selective costimulation modulators (e.g., abatacept), should be avoided.(1) Concomitant use of these biologic agents with tocilizumab has not been studied, and there is a possibility of increased immunosuppression and increased risk of infection with concomitant use.(1) Tocilizumab has been shown to induce clinical responses, improve physical function, and inhibit progression of structural damage in adults with rheumatoid arthritis.(1)

Although use of tocilizumab is not FDA-approved for treatment-naive patients with rheumatoid arthritis,(1) several studies have been published demonstrating potential efficacy of IV and subcutaneous tocilizumab for treatment-naive patients( with recent onset rheumatoid arthritis.82)(83)(84)(85)(86)

Clinical Experience with IV Tocilizumab

Safety and efficacy of IV tocilizumab have been evaluated in 5 randomized, double-blind studies in adults (≥18 years of age) with moderate to severe active rheumatoid arthritis.(1)(3)(4)(5)(6)(8) In these studies, tocilizumab was administered as monotherapy, in combination with methotrexate or other nonbiologic DMARDs in patients with an inadequate response to these drugs, or in combination with methotrexate in patients with an inadequate response to TNF blocking agents.(1) Patients included in these studies had 8 or more tender joints and 6 or more swollen joints.(1)(3)(4)(5)(6) Those receiving low stable dosages of corticosteroids (equivalent to 10 mg or less of prednisone daily) and/or stable dosages of nonsteroidal anti-inflammatory agents (NSAIAs) could continue such agents.(3)(4)(5)(6)(8) Tocilizumab was administered IV at a dosage of 4 or 8 mg/kg once every 4 weeks.(1)(3)(4)(5)(6)

The American College of Rheumatology (ACR) criteria for improvement (ACR response) in measures of disease activity were used as the principal measure of clinical response in studies evaluating the efficacy of tocilizumab.(1)(3)(4)(5)(6) An ACR 20 response is achieved if the patient experiences a 20% improvement in tender and swollen joint count and a 20% or greater improvement in at least 3 of the following criteria: patient pain assessment, patient global assessment, physician global assessment, patient self-assessed disability, or laboratory measures of disease activity (i.e., erythrocyte sedimentation rate [ESR] or C-reactive protein [CRP] level).(2) An ACR 70 response is defined using the same criteria but with a level of improvement of 70%.(16)(17)(18) The proportion of patients who achieved an ACR 20 response at week 24 was the primary end point in the studies of tocilizumab.(1)(3)(4)(5)(6) In one longer-term study, the total Sharp-Genant score (a composite score of erosions and joint space narrowing in hands, wrists, and forefeet) was used as the principal measure of joint damage, and the Health Assessment Questionnaire Disability Index (HAQ-DI) was used to assess physical function and disability.(1)

Clinical evaluations of tocilizumab indicate that, in adults with active rheumatoid arthritis despite therapy with methotrexate or other nonbiologic DMARD(s), the addition of tocilizumab to the DMARD regimen is associated with greater efficacy than use of the nonbiologic DMARD(s) alone.(1)(4)(5) In addition, therapy with tocilizumab in conjunction with methotrexate has been more effective than methotrexate alone in patients with an inadequate response to prior therapy with TNF blocking agents.(1)(6) In patients with an inadequate response to prior therapy with nonbiologic DMARDs or TNF blocking agents, response rates were higher in those receiving tocilizumab 8 mg/kg IV every 4 weeks compared with those receiving 4 mg/kg IV every 4 weeks.(1)(4)(6) When tocilizumab was compared with methotrexate as monotherapy in patients with active rheumatoid arthritis, tocilizumab was more effective than methotrexate.(1)(3) Response to tocilizumab can occur 2–4 weeks following initiation of therapy in some patients.(3)(4)(5)(6)(11)

In the study evaluating efficacy of tocilizumab as monotherapy (AMBITION), patients were randomized to receive either tocilizumab (8 mg/kg IV every 4 weeks) or methotrexate (7.5 mg weekly, increased up to 20 mg weekly).(1)(3) Patients were excluded if they had received methotrexate in the past 6 months, had previously discontinued methotrexate because of lack of response or clinically important toxicity, or had previously discontinued therapy with a TNF blocking agent because of inadequate response.(1)(3) About two-thirds of the patients in the study had never received methotrexate therapy.(1)(3) An ACR 20 response was achieved in 70% of patients receiving tocilizumab and 53% of those receiving methotrexate for 24 weeks.(1)(3)

In 2 studies (OPTION and LITHE), therapy with tocilizumab given in combination with methotrexate was evaluated in patients who had not responded adequately to methotrexate; patients in these studies were randomized to receive tocilizumab 4 mg/kg IV every 4 weeks, tocilizumab 8 mg/kg IV every 4 weeks, or placebo, each given in conjunction with a stable dosage of methotrexate (10–25 mg weekly).(1)(4)(29) Patients who had been treated unsuccessfully with a TNF blocking agent were excluded from these studies.(4)(8)(29) In one study (OPTION), ACR 20 responses were achieved at 24 weeks in 59, 48, or 27% of those receiving tocilizumab 8 mg/kg and methotrexate, tocilizumab 4 mg/kg and methotrexate, or placebo and methotrexate, respectively.(1)(4) The other study (LITHE) was longer term (2 years with an optional 3-year extension phase) and was designed to assess changes in signs and symptoms of rheumatoid arthritis at 24 weeks, with subsequent assessments of joint damage and physical functioning at 1 and 2 years of treatment.(1)(8)(29) Following 1 year of treatment with the randomly assigned regimen, patients received open-label treatment with tocilizumab 8 mg/kg for an additional year or had the option to continue their double-blind treatment if improvement in swollen and tender joint count was maintained above 70%.(1) Planned interim analyses indicated that ACR 20 responses were achieved at 24 weeks in 56, 51, or 27% and at 1 year in 56, 47, or 25% of those receiving tocilizumab 8 mg/kg and methotrexate, tocilizumab 4 mg/kg and methotrexate, or placebo and methotrexate, respectively.(1) At 1 year, 7, 4, or 1% of patients receiving tocilizumab 8 mg/kg and methotrexate, tocilizumab 4 mg/kg and methotrexate, or placebo and methotrexate, respectively, had achieved major clinical responses (defined as ACR 70 responses for a continuous 24-week period).(1)(29) Findings at 1 year also indicated that treatment with tocilizumab 4 or 8 mg/kg and methotrexate inhibited progression of structural damage compared with placebo and methotrexate.(1)(29) At 1 year, 78 or 83% of patients receiving tocilizumab 4 or 8 mg/kg and methotrexate, respectively, exhibited no radiographic progression, as determined by change in total Sharp-Genant score, compared with 66% of patients receiving placebo and methotrexate.(1) Patients receiving tocilizumab 4 or 8 mg/kg and methotrexate also experienced greater improvements in physical function and disability, as assessed using HAQ-DI scores, at 1 year compared with those who received placebo and methotrexate.(1)(29) By the end of the 2-year study, most patients in the placebo and methotrexate group had crossed over to tocilizumab and methotrexate treatment.(1)

In one study (TOWARD) in patients who had not responded adequately to treatment with one or more nonbiologic DMARDs, tocilizumab (8 mg/kg IV weekly) or placebo was added to the stable background DMARD regimen.(1)(5) For 76% of patients in the study, the background regimen contained one DMARD, most commonly methotrexate (mean dosage of 15 mg weekly).(5) Patients who had been treated unsuccessfully with a TNF blocking agent or had previously received any cell-depleting therapy were excluded.(5) At 24 weeks, an ACR 20 response was achieved in 61 or 24% of those receiving tocilizumab or placebo, respectively, in conjunction with the nonbiologic DMARD(s).(1)(5)

Tocilizumab, given in combination with methotrexate, also was evaluated in one study (RADIATE) in patients with rheumatoid arthritis who had not responded adequately to, or who had not tolerated, one or more TNF blocking agents.(1)(6) Patients in this study were randomized to receive tocilizumab 4 mg/kg IV every 4 weeks, tocilizumab 8 mg/kg IV every 4 weeks, or placebo, each given in combination with methotrexate 10–25 mg weekly; tocilizumab was initiated following discontinuance of any other DMARDs and stabilization of the methotrexate dosage.(1)(6) At 24 weeks, an ACR 20 response was achieved in 50, 30, or 10% of those receiving tocilizumab 8 mg/kg and methotrexate, tocilizumab 4 mg/kg and methotrexate, or placebo and methotrexate, respectively.(1)(6)

Analysis of long-term data from the 5 studies showed that improvements from baseline in clinical measures of efficacy generally were sustained through at least 4.2 years of tocilizumab therapy.(30)

General health status was assessed by the Short Form Health Survey (SF-36) in these 5 studies.(1) Patients receiving tocilizumab demonstrated greater improvement from baseline compared with placebo in the physical and mental component summaries and in all 8 domains of the SF-36.(1)

A phase 4, multicenter, open-label randomized trial evaluated the efficacy of tocilizumab compared to rituximab in 161 patients with rheumatoid arthritis who demonstrated an inadequate response to TNF-blocking agents.(73) In this study, 161 patients were stratified based on results of synovial B-cell status determined by biopsy, and randomized to receive two 1000-mg rituximab infusions 2 weeks apart or 8 mg/kg tocilizumab monthly infusions; baseline synovial biopsies from all participants were also subjected to RNA sequencing. (73) In patients who were histologically classified as B-cell rich, no statistically significant difference between treatments was observed in the number of patients who achieved the primary outcome (50% improvement from baseline in the Clinical Disease Activity Index [CDAI]).(73) However, in patients who were classified as having poor B-cell status based on RNA sequencing, a significantly higher response rate for the primary outcome was observed in those who received tocilizumab (63%) than those who received rituximab (36%).(73)

Clinical Experience with Subcutaneous Tocilizumab

Efficacy and safety of subcutaneous tocilizumab have been evaluated in 2 double-blind, controlled studies in adults with moderate to severe active rheumatoid arthritis.(1)(22)(23) One study was a noninferiority study that compared tocilizumab 162 mg subcutaneously every week with tocilizumab 8 mg/kg IV every 4 weeks.(1)(23) The other study was a placebo-controlled superiority study evaluating tocilizumab 162 mg subcutaneously every other week.(1)(22) In these studies, the effects of subcutaneously administered tocilizumab on clinical response and progression of joint damage were consistent with the effects observed in studies evaluating IV administration of the drug.(1)

In the active-control study (SUMMACTA), 1262 patients with moderate to severe active rheumatoid arthritis were randomized in a 1:1 ratio to receive subcutaneous tocilizumab (162 mg every week) or IV tocilizumab (8 mg/kg every 4 weeks) for 24 weeks in conjunction with a stable dosage of one or more nonbiologic DMARDs.(1)(23) To be included in the study, patients were required to have at least 4 tender and 4 swollen joints and to have had an inadequate response to at least one DMARD.(1)(23) Patients receiving stable dosages of NSAIAs and/or low stable dosages of corticosteroids (equivalent to 10 mg or less of prednisone daily) could continue such agents.(23) Approximately 21% of patients included in the study had experienced an inadequate response to prior TNF-blocking agent therapy.(23) ACR 20 response at 24 weeks (the primary measure of efficacy) was observed in 69 or 73% of patients receiving subcutaneous or IV tocilizumab, respectively, establishing noninferiority of the subcutaneous regimen.(1)(23) ACR 50 and ACR 70 responses were observed in 47 and 24%, respectively, of those receiving subcutaneous tocilizumab and 49 and 28%, respectively, of those receiving IV tocilizumab.(1)(23) At 24 weeks, similar proportions of patients receiving subcutaneous or IV tocilizumab (38 or 37%, respectively) had a low level of disease activity, as defined by a Disease Activity Score in 28 joints (DAS 28) of less than 2.6.(1)(23) The mean decrease in HAQ-DI score from baseline to week 24 was 0.6 in both treatment groups, and similar proportions of patients receiving subcutaneous or IV tocilizumab (65 or 67%, respectively) achieved clinically important improvement in HAQ-DI score (change from baseline of at least 0.3 units).(1)(23)

Following the 24-week double-blind period in the SUMMACTA study, patients who had received subcutaneous tocilizumab were randomized in an 11:1 ratio to receive subcutaneous or IV tocilizumab, while those who had received IV tocilizumab were randomized in a 2:1 ratio to receive IV or subcutaneous tocilizumab in a 72-week open-label extension of the study.(24) The proportions of patients who achieved ACR responses, DAS 28 score indicating a low level of disease activity, and clinically important improvement in HAQ-DI score were maintained through week 72 (97 weeks of tocilizumab treatment) and were similar across treatment groups.(24)

In the placebo-controlled study (BREVACTA), 656 patients with moderate to severe active rheumatoid arthritis were randomized in a 2:1 ratio to receive subcutaneous tocilizumab (162 mg every other week) or placebo for 24 weeks in conjunction with a stable dosage of one or more nonbiologic DMARDs.(1)(22) To be included in the study, patients were required to have at least 8 tender and 6 swollen joints and to have had an inadequate response to at least one DMARD.(1)(22) Patients receiving stable dosages of NSAIAs and/or low stable dosages of corticosteroids (equivalent to 10 mg or less of prednisone daily) could continue such agents.(22) Approximately 21% of patients included in the study had experienced an inadequate response to prior TNF-blocking agent therapy.(22) ACR 20 response at 24 weeks (the primary measure of efficacy) was observed in 61% of patients receiving tocilizumab compared with 32% of those receiving placebo.(1)(22) In addition, ACR 50 and ACR 70 responses were observed at 24 weeks in 40 and 20%, respectively, of tocilizumab-treated patients compared with 12 and 5%, respectively, of placebo recipients.(1)(22) At 24 weeks, 32 or 4% of patients receiving tocilizumab or placebo, respectively, had a low level of disease activity, as defined by a DAS 28 score of less than 2.6.(1)(22) Less progression of joint damage, as assessed radiographically by the change in van der Heijde-modified Sharp score (a composite score of structural damage that measures joint erosions and joint space narrowing in the hands, wrists, and feet(25) ), was observed in patients receiving tocilizumab compared with those receiving placebo.(1)(22) The mean decrease in HAQ-DI score from baseline to week 24 was 0.4 or 0.3, and the proportion of patients who achieved clinically important improvement in HAQ-DI score (change from baseline of at least 0.3 units) was 58 or 47% for patients receiving tocilizumab or placebo, respectively.(1) In an open-label continuation of this study, efficacy and safety of tocilizumab were maintained for up to 2 years in patients receiving tocilizumab every 2 weeks.(74) Escalation to weekly dosing demonstrated response and tolerability in prior nonresponders.(74)

In the SUMMACTA study, the proportion of patients achieving responses (ACR responses and DAS 28 scores indicating low disease activity) to either IV tocilizumab (8 mg/kg every 4 weeks) or subcutaneous tocilizumab (162 mg every week) generally were similar regardless of body weight, but tended to be lower and more variable in the small subset of patients weighing 100 kg or more.(23) However, in the BREVACTA study, patients weighing 100 kg or more had poorer ACR responses to subcutaneous tocilizumab (162 mg every other week) than did patients in 2 lower-weight categories.(22)

Other randomized clinical studies have also demonstrated efficacy of weekly dosing of subcutaneous tocilizumab in patients with inadequate response to every-other-week dosing.(75)(76) Additionally, maintenance of efficacy has been shown in studies of tocilizumab alone (after discontinuation of methotrexate) in patients with low disease activity on methotrexate plus tocilizumab.(77)(78)

Clinical Perspective

The American College of Rheumatology issued guidelines for the treatment of rheumatoid arthritis in 2021.(2003) Recommendations for disease-modifying treatments for rheumatoid arthritis include conventional DMARDs (e.g., hydroxychloroquine, leflunomide, methotrexate, sulfasalazine), biologic DMARDs (e.g., TNF blocking agents, abatacept, tocilizumab, sarilumab, rituximab), and/or targeted synthetic DMARDs (e.g., Janus kinase inhibitors).(2003) Specific agents for rheumatoid arthritis treatment are selected according to current disease activity, prior therapies used, and the presence of comorbidities.(2003) A “treat-to-target” approach is typically employed, with the goal of achieving low disease activity or remission.(2003)

IL-6 inhibitors used in the treatment of rheumatoid arthritis include tocilizumab and sarilumab.(2003) Methotrexate monotherapy is strongly recommended over biologic DMARD monotherapy for DMARD-naive patients with moderate to high disease activity, because of its established safety and efficacy and lower cost.(2003) Add-on therapy with biologic DMARDs (including IL-6 inhibitors) and targeted synthetic DMARDs is conditionally recommended over triple therapy (i.e., addition of sulfasalazine and hydroxychloroquine) for patients who are taking maximally tolerated doses of methotrexate and are not at target.(2003) Recommendations for the use and selection of biologic DMARDs in rheumatoid arthritis vary based on the presence of certain comorbidities (e.g., heart failure, previous serious infection, nontuberculous mycobacterial lung disease).(2003)

Juvenile Idiopathic Arthritis

Tocilizumab is used for the treatment of active systemic or polyarticular juvenile idiopathic arthritis (JIA) in patients ≥2 years of age.(1)(20)(21) Tocilizumab can be used alone or in combination with methotrexate.(1) Concomitant use of tocilizumab with other biologic DMARDs should be avoided.(1) Such concomitant use has not been studied, and there is a possibility of increased immunosuppression and increased risk of infection with concomitant use.(1) Tocilizumab has been shown to produce clinical improvement in patients 2–17 years of age with active polyarticular JIA.(1)(20) Tocilizumab also has been shown to produce clinical improvement and improve physical function in patients 2–17 years of age with active systemic JIA.(1)(21)

Clinical Experience with IV Tocilizumab in Polyarticular Juvenile Idiopathic Arthritis

IV tocilizumab has been evaluated in a randomized, double-blind treatment-withdrawal study (CHERISH) in patients 2–17 years of age with active polyarticular JIA who had not responded adequately to or had not tolerated treatment with methotrexate.(1)(20) Patients included in this study had at least 6 months of active disease, with at least 5 joints with active arthritis (swollen or limitation of movement with pain and/or tenderness) and limitation of motion in at least 3 of the active joints.(1)(20) Disease subtypes included rheumatoid factor-positive or -negative polyarticular JIA and extended oligoarticular JIA.(1)(20) Patients receiving low stable dosages of corticosteroids (equivalent to 0.2 mg/kg or less [maximum of 10 mg] of prednisone daily) and/or a stable dosage of NSAIAs or methotrexate (10–20 mg/m2 per week) could continue such therapy;(1)(20) however, concomitant use of other nonbiologic or biologic DMARDs was not permitted.(1) Approximately 32% of patients had received prior biologic DMARD therapy.(20)

During a 16-week lead-in period, all 188 patients enrolled in the study received IV tocilizumab; those weighing 30 kg or more received a dosage of 8 mg/kg every 4 weeks, while those weighing less than 30 kg were randomized to receive a dosage of either 8 or 10 mg/kg every 4 weeks.(1)(20) At week 16, all patients with JIA-ACR 30 response were randomly assigned in a 1:1 ratio (stratified by concomitant methotrexate and corticosteroid use) to continue receiving tocilizumab at the previously assigned dosage or to receive placebo for 24 weeks.(1)(20) JIA-ACR responses are defined as the percentage improvement (e.g., 30, 50, 70, or 90%) in 3 of any 6 core outcome variables compared with baseline, with worsening in no more than 1 of the remaining variables by 30% or more.(1)(20) Core outcome variables consist of physician global assessment, parent or patient global assessment, number of joints with active arthritis, number of joints with limitation of movement, erythrocyte sedimentation rate (ESR), and physical function (as assessed using the disability index of the Childhood Health Assessment Questionnaire [CHAQ-DI]).(1)(21)

At the end of the lead-in period, 91% of patients receiving tocilizumab in combination with methotrexate and 83% of those receiving tocilizumab monotherapy (a total of 163 patients) achieved a JIA-ACR 30 response and entered the treatment-withdrawal period; JIA-ACR 50 and JIA-ACR 70 responses were observed at the end of the lead-in period in 84 and 64%, respectively, of patients receiving tocilizumab in combination with methotrexate and 80 and 55%, respectively, of those receiving tocilizumab monotherapy.(1) Response rates for children weighing less than 30 kg and receiving the 8-mg/kg dose were numerically lower than those for the other 2 treatment groups.(20)

During the treatment-withdrawal period, a greater proportion of patients receiving placebo (48%) compared with those receiving tocilizumab (26%) experienced a JIA-ACR 30 flare (the primary measure of efficacy).(1)(20) JIA-ACR 30 flare was defined as 3 or more of the 6 core outcome variables worsening by at least 30% with no more than 1 of the remaining variables improving by more than 30% relative to the start of the treatment-withdrawal period.(1)(20) At the end of the treatment-withdrawal evaluation period, JIA-ACR 30, 50, and 70 responses were observed in a greater proportion of patients receiving tocilizumab compared with those receiving placebo.(1)

Follow-up of patients in a subsequent open-label extension period is ongoing.(1)(20)

Clinical Experience with Subcutaneous Tocilizumab in Polyarticular Juvenile Idiopathic Arthritis

Subcutaneous tocilizumab was evaluated in pediatric patients with polyarticular JIA in a 52-week, open-label, multicenter, pharmacokinetic, pharmacodynamic, and safety study to determine the appropriate subcutaneous dosage of tocilizumab that achieves comparable pharmacokinetic/pharmacodynamic profiles as the IV tocilizumab regimen.(1) Polyarticular JIA patients 1–17 years of age with an inadequate response to or inability to tolerate methotrexate, including patients with well-controlled disease on IV tocilizumab therapy and tocilizumab-naive patients with active disease, were treated with subcutaneous tocilizumab based on their body weight.(1) Patients weighing 30 kg or more (25 patients) received 162 mg of tocilizumab given subcutaneously every 2 weeks and patients weighing less than 30 kg (27 patients) received 162 mg of tocilizumab subcutaneously every 3 weeks for 52 weeks.(1) Of these 52 patients, 71% were naive to tocilizumab and 29% had been receiving IV tocilizumab and were switched to subcutaneous tocilizumab at baseline.(1) The manufacturer states that the efficacy of subcutaneous tocilizumab in pediatric patients 2–17 years of age is based on pharmacokinetic exposure and extrapolation of the established efficacy of IV tocilizumab in pediatric patients with polyarticular JIA and subcutaneous tocilizumab in adults with rheumatoid arthritis.(1)

Clinical Experience with IV Tocilizumab in Systemic Juvenile Idiopathic Arthritis

Tocilizumab has been evaluated in a 12-week randomized, double-blind, placebo-controlled study in 112 patients 2–17 years of age with active systemic JIA that had not responded adequately to treatment with NSAIAs and corticosteroids.(1)(21) Patients included in this study had at least 6 months of active disease, with either 5 or more active joints or 2 or more active joints and fever.(21) Patients were randomized in a 2:1 ratio to receive tocilizumab (administered IV every 2 weeks at a weight-adjusted dosage [8 mg/kg in those weighing 30 kg or more, 12 mg/kg in those weighing less than 30 kg]) or placebo and were stratified according to body weight, disease duration, corticosteroid dosage, and methotrexate use.(1)(21) Those receiving stables dosages of NSAIAs, corticosteroids (equivalent to 0.5 mg/kg or less [maximum of 30 mg] of prednisone daily), and methotrexate (20 mg/m2 or less per week) could continue such therapy; however, concomitant use of other nonbiologic or biologic DMARDs was not permitted.(21) Approximately 82% of patients had received prior biologic DMARD therapy.(21) Tapering of the corticosteroid dosage was permitted beginning at week 6 in patients who achieved a JIA-ACR 70 response.(1)(21) At week 12, the combined primary end point of JIA-ACR 30 response and absence of fever was observed in 85% of patients receiving tocilizumab compared with 24% of those receiving placebo.(1)(21) JIA-ACR 30, 50, and 70 responses were observed at week 12 in 91, 85, and 71%, respectively, of patients receiving tocilizumab, compared with 24, 11, and 8%, respectively, of those receiving placebo.(1)(21) Tocilizumab therapy also was associated with improvement in systemic manifestations (fever and rash).(1)(21) A greater proportion of patients receiving tocilizumab reduced their corticosteroid dosage by at least 20% without experiencing a subsequent occurrence of systemic symptoms or JIA-ACR 30 flare, compared with placebo recipients (24 versus 3%).(1) In addition, a greater proportion of patients receiving tocilizumab had clinically important improvements in physical function, as assessed using the CHAQ-DI, from baseline to week 12, compared with those receiving placebo (77 versus 19%).(1)

A total of 110 patients subsequently were entered into a long-term, single-group, open-label extension study.(21) At 44 weeks of follow-up, responses to tocilizumab therapy (absence of fever, JIA-ACR response rates, systemic symptoms) were similar to those observed in the 12-week randomized study.(1) By week 44, 43% of patients had discontinued oral corticosteroid therapy; about 50% of these patients had discontinued corticosteroid use for 18 weeks or longer.(1) At 52 weeks of follow-up, 80% of patients had JIA-ACR 70 response and no fever, 59% had JIA-ACR 90 response and no fever, 48% had no joints with active arthritis, and 52% had discontinued oral corticosteroid therapy; the proportion of patients with moderate or severe functional impairment was 38%, compared with 82% at study baseline.(21)

Clinical Experience with Subcutaneous Tocilizumab in Systemic Juvenile Idiopathic Arthritis

Subcutaneous tocilizumab was evaluated in pediatric patients with systemic JIA in a 52-week, open-label, multicenter, pharmacokinetic, pharmacodynamic, and safety study to determine the appropriate subcutaneous dosage of tocilizumab that achieves comparable pharmacokinetic/pharmacodynamic profiles as the IV tocilizumab regimen.(1) Eligible patients received tocilizumab subcutaneously dosed according to body weight, with patients weighing 30 kg or more (26 patients) treated with 162 mg of tocilizumab every week and patients weighing less than 30 kg (25 patients) treated with 162 mg of tocilizumab every 10 days (8 patients) or every 2 weeks (17 patients) for 52 weeks.(1) Of these patients, 51% were naive to subcutaneous tocilizumab and 49% had been receiving IV tocilizumab and were switched to subcutaneous tocilizumab at baseline.(1) Efficacy of subcutaneous tocilizumab in pediatric patients 2–17 years of age is based on pharmacokinetic exposure and extrapolation of the established efficacy of IV tocilizumab in pediatric patients with systemic JIA.(1)

Clinical Perspective

The American College of Rheumatology and the Arthritis Foundation issued a joint guideline in 2019 for the treatment of juvenile idiopathic arthritis manifesting as nonsystemic polyarthritis (including polyarticular disease), sacroiliitis, or enthesitis.(2013) Several drug classes are used to treat juvenile idiopathic arthritis, including NSAIAs, systemic and intra-articular corticosteroids, conventional DMARDs (e.g., methotrexate, sulfasalazine, hydroxychloroquine, leflunomide), and biologic DMARDs (e.g., TNF blocking agents, abatacept, tocilizumab, rituximab).(2013) Specific agents for juvenile idiopathic arthritis treatment are selected according to the presence of certain risk factors (e.g., positive anti-cyclic citrullinated peptide antibodies, positive rheumatoid factor, joint damage), level of disease activity, involvement of specific joints, presence of certain comorbidities (e.g., uveitis), and prior therapies received.(2013)(2022) An individualized “treat-to-target” approach is typically employed, with the goal of achieving remission or minimal/low disease activity.(2014)

Biologic DMARDs, including tocilizumab, are conditionally recommended as an initial therapy option in patients with risk factors and involvement of high-risk joints such as the cervical spine, wrist, or hip; high disease activity; and/or those at high risk of disabling joint disease.(2013) Use of tocilizumab is also conditionally recommended as subsequent therapy in patients with moderate/high disease activity as an add-on therapy to a DMARD or when switching from a TNF blocking agent.(2013) In patients receiving a second biologic DMARD, the guideline conditionally recommends tocilizumab, abatacept, or a TNF blocking agent over rituximab.(2013)

Giant Cell Arteritis

Tocilizumab is used for the management of giant cell arteritis (GCA) in adults.(1)(50)(51)(52)(53) The drug can be used in combination with a tapering course of corticosteroids or alone following discontinuance of corticosteroids.(1)

Clinicians treating patients with GCA should be aware that monitoring CRP concentrations is not a reliable method to detect active disease or relapse of GCA in patients who are receiving IL-6 inhibitors such as tocilizumab since they may suppress CRP levels regardless of disease activity.(51)(52)

Clinical Experience with Subcutaneous Tocilizumab

Efficacy and safety of subcutaneous tocilizumab in the treatment of GCA were demonstrated in a randomized, double-blind, multicenter study of 1 year's duration (the Giant-Cell Arteritis Actemra [GiACTA] trial; NCT01791153) in adults with active GCA.(1)(50) In this study, 251 patients with new-onset or relapsing GCA were randomized in a 2:1:1:1 ratio to one of four treatment arms; 2 subcutaneous dosages of tocilizumab (162 mg every week and 162 mg every other week) with a prespecified 26-week prednisone taper regimen were compared with 2 different placebo control groups (prespecified prednisone taper regimen over 26 weeks and 52 weeks).(1)(50) The study consisted of a 52-week, blinded period followed by a 104-week, open-label extension.(1)(50) All patients concurrently received corticosteroid therapy (i.e., prednisone).(1) Both tocilizumab treatment groups and one of the placebo groups followed a prespecified prednisone taper regimen with the aim to reach 0 mg by 26 weeks, while the second placebo group followed a prespecified prednisone taper regimen with the aim to reach 0 mg by 52 weeks, which was designed to be more in line with standard clinical practice.(1)(50) The primary efficacy end point was the proportion of patients achieving sustained corticosteroid-free remission at 52 weeks.(1)(50) Sustained remission was defined by a patient attaining a sustained absence of GCA signs and symptoms from week 12 through week 52, normalization of the erythrocyte sedimentation rate (ESR) from week 12 through week 52, normalization of CRP from week 12 through week 52, and successful adherence to the prednisone taper defined by not more than 100 mg of excess prednisone from week 12 through week 52.(1)(50)

In the GiACTA trial (NCT01791153), tocilizumab 162 mg weekly and 162 mg every other week plus a 26-week prednisone taper both showed superiority in achieving sustained remission from week 12 through week 52 (56 and 53% of patients, respectively) compared with placebo plus a 26-week prednisone taper (14%) or placebo plus a 52-week prednisone taper (18%).(1)(50) In addition, the estimated annual cumulative prednisone dose was lower in the 2 tocilizumab treatment groups (medians of 1887 and 2207 mg in the once-weekly and every-2-week tocilizumab treatment groups, respectively) compared with the placebo groups (medians of 3804 and 3902 mg in the placebo plus 26-week prednisone taper and the placebo plus 52-week prednisone taper groups, respectively).(1) The overall tolerability profile of tocilizumab in the treatment of GCA was found to be similar to its tolerability profile in other indications.(1)(50) A post-hoc analysis of this study demonstrated sustained remission and a reduction in flares with tocilizumab compared to placebo in subgroups of patients who had either cranial symptoms only, polymyalgia rheumatica symptoms only, or a combination of both at baseline evaluation.(79)

In a 2-year extension study (part 2) of GiACTA, data from 215 of the original 251 randomized patients were analyzed.(53) Participants who were in clinical remission at the end of the 52-week, double-blind period received no further treatment and those not in clinical remission received therapy at the clinician's discretion, which could include tocilizumab and/or corticosteroids.(53) A higher proportion of patients who were originally assigned to receive tocilizumab were treatment-free during this period (with a higher proportion of treatment-free patients among those who received the drug once weekly compared with every 2 weeks) compared with those originally assigned to receive placebo.(53) Retreatment with tocilizumab with or without corticosteroids restored clinical remission in patients who experienced a disease flare.(53) In addition, cumulative corticosteroid doses were lower in patients originally assigned to tocilizumab than in patients originally assigned to placebo.(53) No new safety concerns emerged in the tocilizumab-treated patients during the 3-year study.(53)

Clinical Experience with IV Tocilizumab

IV tocilizumab for the treatment of GCA was assessed in an open-label pharmacokinetic-pharmacodynamic (PK-PD) and safety study (WP41152; NCT03923738) to determine the appropriate IV dose of tocilizumab that would achieve comparable PK-PD profiles to subcutaneous tocilizumab.(1)(93) A total of 24 patients who had received ≥5 doses of tocilizumab 8 mg/kg IV every 4 weeks and achieved remission were enrolled in the study.(1)(93) In period 1 of the study, all patients received tocilizumab 7 mg/kg IV every 4 weeks for 20 weeks.(1)(93) After completion of period 1, 22 patients remained in remission and continued in period 2 to receive tocilizumab 6 mg/kg IV every 4 weeks for 20 weeks.(1)(93) Both dosages of tocilizumab were generally well tolerated.(93) The efficacy of IV tocilizumab 6 mg/kg in adults with GCA is based on pharmacokinetic exposure and extrapolation to the efficacy established for subcutaneous tocilizumab in patients with GCA.(1)

Clinical Perspective

Giant cell arteritis (GCA) is a systemic vasculitis that primarily involves medium-sized and large arteries; the condition can cause a variety of clinical manifestations, including headaches, ischemic visual symptoms (with a risk of vision loss), limb or jaw claudication, polymyalgia rheumatica, aortic aneurysm, myocardial infarction, and stroke.(50)(51)(52)(3000) The condition mainly occurs in adults older than 50 years of age and is 2–3 times more likely to occur in women than in men.(50)(51)(52) Corticosteroids are considered standard first-line initial therapy for GCA and can help control headaches and reduce systemic inflammation, normalize inflammatory markers such as C-reactive protein (CRP), and prevent vision loss.(50)(51)(52) However, long-term corticosteroid therapy can cause severe adverse effects in patients with GCA.(50)(51)

Because concentrations of CRP and other acute-phase reactants increase with elevated serum concentrations of interleukin-6 (IL-6), tocilizumab has been studied as a treatment for GCA.(50)(51)(52)(53) Clinical experience with tocilizumab in GCA to date indicates that the drug can produce sustained remission and allow for reductions in corticosteroid doses (i.e., steroid-sparing effect) that are used to control the disease and to maintain remission.(50)(51)(52)(53) Further study is needed to determine the optimal role of tocilizumab therapy in the management of GCA and to evaluate its efficacy when used alone without corticosteroids.(50)(51)(52) Tocilizumab has been shown to be an effective glucocorticoid-sparing agent for GCA.(3000) Therefore, some clinicians recommend that tocilizumab therapy be considered in patients who are experiencing adverse effects from corticosteroid therapy, patients with clinically important preexisting medical conditions that could be adversely affected by long-term corticosteroid therapy (e.g., diabetes mellitus, hypertension, psychiatric conditions, pancreatitis), and patients whose disease is refractory to corticosteroid therapy (i.e., patients who didn't achieve remission or who experienced flares during a corticosteroid taper).(51)(52)

The American College of Rheumatology and Vasculitis Foundation issued a joint guideline in 2021 for the management of GCA.(3000) Medications used to treat GCA include systemic glucocorticoids, non-biologic immunosuppressants (e.g., azathioprine, leflunomide, methotrexate, mycophenolate mofetil, cyclophosphamide), and biologic DMARDs (e.g., TNF blocking agents, abatacept, tocilizumab).(3000) Specific drug regimens are recommended according to the degree of organ involvement.(3000) The guideline conditionally recommends the use of tocilizumab with oral glucocorticoids over oral glucocorticoids alone in patients with newly diagnosed GCA.(3000) A conditional recommendation is also given for adding tocilizumab and increasing the dose of glucocorticoids over adding methotrexate and increasing the dose of glucocorticoids in patients who experience disease relapse with cranial symptoms while receiving glucocorticoids alone.(3000) Choice of therapy should be based on the physician’s experience and the patient’s clinical condition, values, and preferences.(3000)

Cytokine Release Syndrome Associated with Chimeric Antigen Receptor T-cell Therapy

Tocilizumab is used in the management of chimeric antigen receptor (CAR) T cell-induced severe or life-threatening cytokine release syndrome (CRS) in adults and pediatric patients ≥2 years of age.(1) Tocilizumab can be used alone or in combination with corticosteroids in the treatment of this condition.(1) CRS is a clinically important adverse effect of CAR T-cell therapies used in the treatment of malignancies.(61) Clinical manifestations may include fever, fatigue, coagulopathy, nausea, capillary leak, and multiorgan dysfunction.(61)

Efficacy of IV tocilizumab in the treatment of CRS was assessed in a retrospective analysis of pooled outcome data from clinical trials of CAR T-cell therapies for hematologic malignancies.(1) Evaluable patients had been treated with tocilizumab 8 mg/kg (12 mg/kg for patients weighing less than 30 kg) with or without additional high-dose corticosteroids for severe or life-threatening CRS; only the first episode of CRS was included in the analysis.(1) The study population included 24 males and 21 females (total 45 patients) of a median age of 12 years (range: 3–23 years); 82% were Caucasian.(1) The median time from the onset of CRS to the first dose of tocilizumab was 4 days (range: 0–18 days).(1) Resolution of CRS was defined as lack of fever and off of vasopressors for at least 24 hours.(1) Patients were considered responders if CRS resolved within 14 days of the first dose of tocilizumab, if no more than 2 doses of tocilizumab were needed, and if no drugs other than tocilizumab and corticosteroids were used for treatment.(1) Thirty-one patients (69%) achieved a response.(1) Achievement of resolution of CRS within 14 days was confirmed in a second study using an independent cohort that included 15 patients (range: 9–75 years old) with CAR T cell-induced CRS.(1)

Clinical Perspective

The American Society of Clinical Oncology (ASCO) issued a guideline in 2021 for the management of immune-related adverse events in patients treated with CAR T-cell therapy. (3001) Specific agents are selected according to grading of fever, hypoxia, and hypotension based on the American Society of Transplantation and Cellular Therapy (ASTCT) consensus grading system.(3001) Management of CRS begins with supportive care (e.g., antipyretics, IV hydration, broad-spectrum antibiotics, granulocyte-colony stimulating factor), but may require pharmacologic treatment for prolonged or severe CRS.(3001) Tocilizumab with or without a corticosteroid is recommended in patients with prolonged or severe CAR T-cell-associated CRS.(3001)

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

Tocilizumab is used for the management of adults with systemic sclerosis-associated interstitial lung disease to slow the rate of pulmonary function decline.(1)(80)(81)

Clinical Experience with Subcutaneous Tocilizumab

Efficacy of tocilizumab in SSc-ILD was evaluated in a phase 3, multicenter, randomized, double-blind, placebo-controlled study (focuSSced) in 212 adults with SSc as defined by the 2013 ACR/EULAR criteria with onset of disease of 5 years or less, modified Rodnan Skin Score (mRSS) of 10–35 units at screening, increased inflammatory markers (or platelets), and active disease.(1)(80) Criteria for active disease included at least one of the following: disease duration 18 months or less, increase in mRSS of 3 or more units over 6 months, involvement of one new body area and an increase in mRSS of at least 2 units over 6 months, or involvement of 2 new body areas within the previous 6 months, or presence of at least one tendon friction rub.(1)(80) Use of biologic agents, alkylating agents, or cyclophosphamide was not permitted.(1)(80)

Patients were randomized to tocilizumab 162 mg or placebo administered weekly by subcutaneous injection for 48 weeks, followed by open-label tocilizumab 162 mg administered subcutaneously every week for another 48 weeks.(1)(80) Rescue treatment was allowed after 16 weeks if there was more than a 10% decline in predicted forced vital capacity (FVC) or after 24 weeks for worsening skin fibrosis.(1)(80) The primary efficacy endpoint was change in mRSS from baseline to week 48.(1)(80) Change from baseline in FVC at week 48 was a secondary endpoint.(1)(80)

Sixty-seven percent of patients in the tocilizumab group and 64% in the placebo group had SSc-ILD.(1)(80) Primary and secondary endpoints were evaluated in the overall population and a post-hoc analysis evaluated outcomes in the subgroup of patients with and without SSc-ILD.(1)(80) No statistically significant difference in the primary outcome of change in mRSS was observed in the overall population or in the subgroup of patients with SSc-ILD.(1)(80) The change from baseline in predicted FVC and observed FVC was substantially improved in the overall population and in SSc-ILD subgroup.(1)(80)

A phase 2/3 multicenter, randomized, double-blind, placebo-controlled study provides supportive information on the efficacy of tocilizumab in adult patients with SSc.(1)(81) Results in terms of mRSS and FVC outcomes were similar to those observed in the principal phase 3 study.(1)(81)

Coronavirus Disease 2019 (COVID-19)

Tocilizumab is used in the management of coronavirus disease 2019 (COVID-19) in hospitalized adult patients who are receiving systemic corticosteroids and require supplemental oxygen, noninvasive or invasive mechanical ventilation, or extracorporeal membrane oxygenation (ECMO).(1) Tocilizumab is also used in the management of COVID-19 in hospitalized pediatric patients 2–18 years of age.(70)

The rationale for tocilizumab's use in this infection is related to its specificity for the interleukin-6 (IL-6) receptor, which may help to relieve symptoms of CRS (e.g., fever, organ failure, death) in severely ill patients with COVID-19.(31)(32)(33)(37)(40)(41)(45)

Clinical Experience with IV Tocilizumab

Efficacy of tocilizumab for the treatment of COVID-19 was evaluated in a randomized, controlled, open-label, multicenter study (RECOVERY; NCT04381936) in 4116 hospitalized adult patients with severe COVID-19-related pneumonia.(1)(89) The primary efficacy endpoint was time to death through day 28.(1)(89) Patients were randomized to receive either standard of care or IV tocilizumab at a weight-tiered dosing comparable to the recommended dosage in addition to standard of care.(1)(89) At baseline, 0.2% of patients were not on supplemental oxygen, 45% of patients required low flow oxygen, 41% required non-invasive ventilation or high-flow oxygen, and 14% required invasive mechanical ventilation.(1)(89) Initially, 82% of patients were reported to be receiving systemic corticosteroids.(1) Tocilizumab improved survival and clinical outcomes at 28 days.(89) In patients who received tocilizumab, the mortality rate was 31% compared with a mortality rate of 35% in those who received standard of care.(1)(89) Reduction in mortality was observed in a larger proportion of patients receiving systemic corticosteroids compared with those not receiving a systemic corticosteroid at randomization; however, this may have been a chance finding.(89) Patients receiving tocilizumab were also more likely to be discharged from the hospital within 28 days and less likely to require invasive mechanical ventilation.(89)

Efficacy of tocilizumab for the treatment of COVID-19 was additionally supported in a randomized, double-blind, placebo-controlled, multicenter study (EMPACTA; NCT04372186) in 377 hospitalized patients with confirmed COVID-19-related pneumonia who were not receiving mechanical ventilation; the study enrolled a substantial proportion of patients from high-risk and racial and ethnic minority groups.(1)(90) The primary efficacy endpoint was time to progression to mechanical ventilation or death through day 28.(1)(90) At baseline, 9% of patients were not on supplemental oxygen, 64% of patients required low flow oxygen, 27% of patients required high-flow oxygen, and 73% were on corticosteroids.(1)(90) Patients were randomized to receive standard of care plus 1 or 2 doses of IV tocilizumab 8 mg/kg (maximum dose 800 mg) or placebo.(90) The cumulative proportion of patients who required mechanical ventilation or died by day 28 was 12% in those receiving tocilizumab and 19.3% in those receiving placebo.(1)(90) Mortality from any cause at day 28 was 10.4% in those receiving tocilizumab compared to 8.6% in those receiving placebo.(1)(90)

In a phase 3 randomized, double-blind, placebo-controlled, multicenter study (COVACTA trial; NCT04320615), 452 adults hospitalized with severe COVID-19-related pneumonia were randomized 2:1 to receive either a single dose of tocilizumab 8 mg/kg or placebo.(1)(91) The primary efficacy end point was improved clinical status, which was measured using a 7-point ordinal scale to assess clinical status based on the need for intensive care and/or ventilator use and the requirement for supplemental oxygen over a 4-week period.(1)(91) The trial failed to meet its primary end point or several key secondary end points.(1)(91) Key secondary end points included 4-week mortality.(91) Differences in the primary end points between the tocilizumab and placebo groups were not statistically significant.(1)(91) At week 4, mortality rates did not differ between the tocilizumab and placebo groups (19.7 versus 19.4%, respectively).(1)(91)

In a double-blind, placebo-controlled, multicenter study (REMDACTA; NCT04 409262). 649 hospitalized patients with severe COVID-19 pneumonia were randomized 2:1 to receive IV tocilizumab and remdesivir or placebo and remdesivir.(1)(92) Tocilizumab was given as a single IV dose of 8 mg/kg along with IV remdesivir 200 mg on day 1 followed by remdesivir 100 mg once daily for a total of 10 days.(1)(92) The primary efficacy endpoint was time from randomization to hospital discharge or “ready for discharge” up to Day 28.(1)(92) At baseline, 7% of patients were on low flow oxygen, 80% were on non-invasive ventilation or high flow oxygen, 14% were on invasive mechanical ventilation, and 84% were on corticosteroids.(1)(92) At day 28, there was no statistically significant difference between the study groups with respect to time to hospital discharge or “ready for discharge” through Day 28.(1)(92) Mortality at day 28 was 18.1% in the tocilizumab and remdesivir study population compared to 19.5% in the placebo and remdesivir study population.(1)(92)

A meta-analysis of the EMPACTA, COVACTA, REMDACTA and RECOVERY studies evaluated the risk difference through Day 28, estimated by the Kaplan-Meier method, in the subgroup of patients receiving baseline corticosteroids.(1) Patients from the RECOVERY trial represent 78.8% of the total sample size in this meta-analysis.(1) The combined risk difference showed that tocilizumab treatment (n=2261) resulted in a 4.61% absolute reduction in the risk of death at Day 28 compared to standard of care.(1)

Emergency Use Authorization in Pediatric Patients

FDA has issued an emergency use authorization (EUA) that permits use of tocilizumab for treatment of COVID-19 in hospitalized pediatric patients 2–18 years of age who are receiving systemic corticosteroids and require supplemental oxygen, non-invasive or invasive mechanical ventilation, or extracorporeal membrane oxygenation (ECMO).(70) FDA states that, based on review of data from an open-label, controlled, platform trial (NCT04381936; RECOVERY); a double-blind, placebo-controlled trial (NCT04320615; COVACTA); a double-blind, placebo-controlled trial (NCT04372186; EMPACTA); and a double-blind, placebo-controlled trial (NCT04409262; REMDACTA]), it is reasonable to believe that tocilizumab may be effective for the treatment of COVID-19 in the pediatric patient population specified in the tocilizumab EUA and, when used under the conditions described in the EUA, the known and potential benefits of tocilizumab when used to treat COVID-19 in such patients outweigh the known and potential risks.(70)  

For additional information about the EUA, the tocilizumab EUA letter of authorization (([Web]),70) EUA fact sheet for healthcare providers (([Web]),71) and EUA fact sheet for patients, parents and caregivers (([Web])72) should be consulted.

For further information on the use of tocilizumab in the treatment of pediatric patients with COVID-19, clinicians also may consult the most recent COVID-19 treatment guidelines from NIH (([Web]).40)

Dosage and Administration

Supplementary dosing info/calc

General

Pretreatment Screening

  • Consider potential risks and benefits of tocilizumab prior to initiating therapy in patients with chronic infection or history of recurrent infection.(1) Do not administer in patients with an active infection until the infection resolves or is appropriately treated.(1)

  • Evaluate for active or inactive tuberculosis prior to initiation of tocilizumab therapy.(1) Do not administer to patients with active tuberculosis.(1)

  • Administer all age-appropriate vaccines prior to starting tocilizumab therapy.(1)

  • Measure absolute neutrophil count (ANC), platelet count, ALT, and AST prior to initiating therapy.(1) In patients with rheumatoid arthritis, giant cell arteritis (GCA), systemic sclerosis-associated interstitial lung disease (SSc-ILD), polyarticular juvenile idiopathic arthritis (PJIA), or systemic juvenile idiopathic arthritis (SJIA), do not initiate therapy if ANC <2000/mm3, platelet count <100,000/mm3, or ALT or AST >1.5 times the upper limit of normal (ULN).(1) In patients with COVID-19, do not initiate therapy if ANC <1000/mm3, platelet count <50,000/mm3, or ALT or AST >10 times ULN.(1)

  • Perform a pregnancy test prior to initiating therapy in females of reproductive potential; if positive, enroll patients in manufacturer’s pregnancy registry.(1)

Patient Monitoring

  • Monitor patients for signs and symptoms of bacterial, viral, or fungal infections. (1)

  • Evaluate for active tuberculosis infection during treatment, even if initial latent tuberculosis test is negative. (1)

  • Monitor patients who present with new-onset abdominal symptoms for signs of GI perforation. (1)

  • Monitor lipid parameters approximately 4 to 8 weeks after start of therapy, and manage according to clinical guidelines.(1)

  • Monitor patients for signs and symptoms of infusion-related and hypersensitivity reactions during and after infusions.(1)

  • Monitor patients for signs and symptoms indicative of demyelinating disorders. (1)

  • In patients with rheumatoid arthritis, GCA, or SSc-ILD: obtain a liver test panel (alkaline phosphatase, total bilirubin, ALT and AST levels) every 4 to 8 weeks after start of therapy for the first 6 months of treatment and every 3 months thereafter.(1)

  • In patients with rheumatoid arthritis, GCA, or SSc-ILD: monitor ANC and platelet count 4 to 8 weeks after start of therapy and every 3 months thereafter.(1)

  • In patients with PJIA: monitor liver test panel (alkaline phosphatase, total bilirubin, ALT and AST levels), ANC, and platelet count at the time of second administration and every 4 to 8 weeks thereafter.(1)

  • In patients with SJIA: monitor liver test panel (alkaline phosphatase, total bilirubin, ALT and AST levels), ANC, and platelet count at the time of second administration and every 2 to 4 weeks thereafter.(1)

Dispensing and Administration Precautions

  • IV infusions of tocilizumab should only be administered by a healthcare professional with appropriate medical support to manage anaphylaxis.(1) If anaphylaxis or other hypersensitivity reaction occurs, stop administration of tocilizumab immediately and permanently discontinue therapy.(1)

Other General Considerations

  • Patients with severe or life-threatening chimeric antigen receptor (CAR) T cell-induced cytokine release syndrome (CRS) frequently have cytopenias or elevated ALT or AST concentrations due to lymphodepleting chemotherapy or the CRS itself.(1) Consider the potential benefit of treating the CRS versus the risks of short-term therapy with the drug.(1)

  • For the management of rheumatoid arthritis, tocilizumab may be used as monotherapy or concomitantly with methotrexate or other nonbiologic disease-modifying antirheumatic drugs (DMARDs).(1) Tocilizumab may be used alone or in combination with methotrexate in patients with systemic or polyarticular JIA.(1)(20)(21)

  • For the management of GCA, administer tocilizumab initially in combination with a tapering course of corticosteroids.(1) Once corticosteroids are discontinued, continue tocilizumab alone.(1)

  • Do not use tocilizumab concomitantly with other biologic DMARDs, such as tumor necrosis factor (TNF) blocking agents, interleukin-1 (IL-1) receptor antagonists, anti-CD20 monoclonal antibodies, and selective costimulation modulators.(1)

Administration

For the management of rheumatoid arthritis, GCA, and systemic or polyarticular JIA, tocilizumab may be administered either by IV infusion over 60 minutes or by subcutaneous injection.(1) The drug should not be administered by rapid IV injection (e.g., IV push or bolus).(1)

For the management of SSc-ILD, tocilizumab is administered by subcutaneous injection.(1) The drug currently is not FDA-labeled for IV administration for this indications.(1) The use of the prefilled pen has not been studied in patients with SSc-ILD.(1)

For the management of CAR T cell-induced CRS, tocilizumab is administered only by IV infusion over 60 minutes.(1) The drug should not be administered by rapid IV injection (e.g., IV push or bolus).(1) Tocilizumab currently is not FDA-labeled for subcutaneous use for this indication.(1)

IV Administration

Tocilizumab injection concentrate is commercially available in single-dose vials (containing 20 mg/mL) and must be diluted prior to IV administration.(1) The appropriate dose of the injection concentrate should be diluted in 0.9 or 0.45% sodium chloride injection to provide a final volume of 50 mL for administration in patients who weigh less than 30 kg and to a final volume of 100 mL for administration in patients who weigh 30 kg or more.(1) A volume of diluent equal to the total required volume of tocilizumab injection concentrate should be removed from the bag or bottle of 0.9 or 0.45% sodium chloride injection prior to the addition of tocilizumab injection concentrate.(1) The total required volume of tocilizumab injection concentrate (0.2, 0.3, 0.4, 0.5, or 0.6 mL/kg for a dose of 4, 6, 8, 10, or 12 mg/kg, respectively) should be added slowly to the diluent, and the bag or bottle should be inverted gently to mix the solution and avoid foaming.(1)

The manufacturer states that tocilizumab infusion solutions are compatible with polypropylene, polyethylene, and polyvinyl chloride infusion bags and polypropylene, polyethylene, and glass infusion bottles.(1) Prior to administration, tocilizumab infusion solutions should be inspected visually for particulate matter and discoloration; if either is present, the solution should be discarded.(1) Tocilizumab infusion solutions using 0.9% sodium chloride injection may be stored at 2–8°C or room temperature for up to 24 hours and should be protected from light.(1) Tocilizumab infusion solutions using 0.45% sodium chloride injection may be stored at 2–8°C for up to 24 hours or at room temperature for up to 4 hours and should be protected from light.(1) Any unused portion remaining in the vial should be discarded since the injection concentrate contains no preservative.(1)

Tocilizumab infusion solutions should be allowed to reach room temperature prior to administration.(1) Tocilizumab should not be infused simultaneously through the same IV line with other drugs.(1)

Subcutaneous Administration

Tocilizumab is administered by subcutaneous injection.(1) Tocilizumab is intended for use under the guidance of a clinician; however, the drug may be self-administered or the patient's caregiver may administer the injection if the clinician determines that the patient and/or their caregiver is competent to safely administer the drug after appropriate training.(1) Polyarticular and systemic JIA patients may self-administer tocilizumab if both the clinician and the parent/legal guardian determine it is appropriate.(1) Patients or caregivers should be instructed to follow the instructions for use for detailed information on administration of the drug.(1)

Tocilizumab for subcutaneous use is commercially available in prefilled single-use syringes or as prefilled single-use autoinjectors.(1) Each syringe or autoinjector delivers 162 mg of tocilizumab in 0.9 mL.(1) The entire contents of the prefilled syringe or autoinjector should be administered as a single dose.(1) Prior to administration, tocilizumab solutions should be inspected visually for particulate matter or discoloration; if the syringe or autoinjector appears to be damaged or the solution contains particulates or is cloudy or discolored, the solution should be discarded.(1)

Tocilizumab injection for subcutaneous use should be protected from light and stored at 2–8°C in the original carton until administration.(1) The injection should not be frozen and the prefilled syringes and autoinjectors should be kept dry.(1) The prefilled syringe or autoinjector should be allowed to sit at room temperature outside of the carton for 30 minutes or 45 minutes, respectively, prior to subcutaneous injection; tocilizumab should not be warmed in any other way (e.g., microwave, hot water).(1)(19)(34) After removal from the refrigerator, the prefilled syringe and autoinjector can be stored up to 2 weeks at or below 30°C.(1) The commercially available subcutaneous injection should not be used for IV administration.(1)

Tocilizumab is administered subcutaneously into the anterior thigh or abdomen (except for the 2-inch area around the umbilicus).(19)(34) Tocilizumab also may be administered subcutaneously into the upper arm by a caregiver.(19)(34) Injection sites should be rotated.(1) Injections should not be made into areas where the skin is tender, bruised, red, hard, or nonintact or into scars or moles.(1)

Dosage

Rheumatoid Arthritis in Adults

For the management of rheumatoid arthritis in adults who have had an inadequate response to one or more DMARDs, the recommended initial dosage of tocilizumab is 4 mg/kg by IV infusion once every 4 weeks; the dosage can be increased to 8 mg/kg by IV infusion once every 4 weeks based on clinical response.(1) Doses exceeding 800 mg per infusion are not recommended.(1)

Alternatively, adults with rheumatoid arthritis who weigh less than 100 kg may receive tocilizumab 162 mg subcutaneously every other week; the dosage can be increased to 162 mg every week based on clinical response.(1) Patients who weigh 100 kg or more may receive tocilizumab 162 mg subcutaneously every week.(1)

When switching from IV to subcutaneous administration, the first subcutaneous dose may be administered in place of the next scheduled IV dose.(1)

Treatment Interruptions or Discontinuance for Toxicity

If certain dose-related laboratory changes (i.e., elevated liver enzyme concentrations, neutropenia, thrombocytopenia) occur in patients with rheumatoid arthritis receiving IV or subcutaneous tocilizumab, interruption of tocilizumab therapy, dose reduction, or discontinuance of therapy may be required ((see Tables 1–3).1)

Juvenile Idiopathic Arthritis

Tocilizumab is administered IV in a weight-adjusted dosage once every 4 weeks in patients with polyarticular JIA and once every 2 weeks in those with systemic JIA.(1) For the management of polyarticular JIA in patients ≥2 years of age, the recommended IV dosage of tocilizumab is 10 mg/kg by IV infusion once every 4 weeks in those weighing <30 kg and 8 mg/kg by IV infusion once every 4 weeks in those weighing 30 kg or more.(1) For the management of systemic JIA in patients ≥2 years of age, the recommended dosage of tocilizumab is 12 mg/kg by IV infusion once every 2 weeks in those weighing <30 kg and 8 mg/kg by IV infusion once every 2 weeks in those weighing ≥30 kg.(1)

Alternatively, tocilizumab may be administered by subcutaneous injection in a weight-adjusted dosage for the management of polyarticular JIA.(1) In patients ≥2 years of age, the recommended subcutaneous dosage of tocilizumab is 162 mg once every 3 weeks in those weighing less than 30 kg and 162 mg once every 2 weeks in those weighing 30 kg or more.(1) For the management of systemic JIA in patients ≥2 years of age, the recommended subcutaneous dosage of tocilizumab is 162 mg once every 2 weeks in those weighing less than 30 kg and 162 mg once weekly in those weighing 30 kg or more.(1)

Because the patient's weight may fluctuate, dosage should not be adjusted based solely on the patient's weight as measured at a single visit.(1) If certain dose-related laboratory changes (i.e., elevated liver enzyme concentrations, neutropenia, thrombocytopenia) occur, interruption of therapy may be required.(1)

When switching from IV to subcutaneous administration, the first subcutaneous dose may be administered in place of the next scheduled IV dose.(1)

Treatment Interruptions or Discontinuance for Toxicity

Tocilizumab dosage reductions have not been evaluated in patients with polyarticular or systemic JIA.(1) However, interruption of tocilizumab therapy is recommended for certain dose-related laboratory changes (i.e., elevated liver enzyme concentrations, neutropenia, thrombocytopenia) at values similar to those considered in adults with rheumatoid arthritis ((see Tables 1–3).1) If clinically appropriate, dosage reduction or discontinuance of concomitant therapy with methotrexate and/or other agents should be considered and tocilizumab withheld pending clinical evaluation.(1) The decision to discontinue tocilizumab therapy because of a laboratory abnormality should be individualized.(1)

Giant Cell Arteritis in Adults

For the management of giant cell arteritis (GCA), the recommended IV dosage of tocilizumab in adults is 6 mg/kg by IV infusion once every 4 weeks in combination with a tapering course of glucocorticoids.(1) Doses >600 mg per infusion are not recommended.(1) Tocilizumab can be used alone following discontinuation of glucocorticoids.(1)

The recommended subcutaneous dosage of tocilizumab in adults with GCA is 162 mg subcutaneously once every week in combination with a tapering course of glucocorticoids.(1) A dosage of 162 mg given once every other week as a subcutaneous injection in combination with a tapering course of corticosteroids also may be prescribed based on clinical considerations.(1) Tocilizumab may be used alone following discontinuance of glucocorticoids.(1)

When switching from tocilizumab IV to subcutaneous administration, the first subcutaneous dose may be administered in place of the next scheduled IV dose.(1)

Treatment Interruptions or Discontinuance for Toxicity

If certain dose-related laboratory changes (i.e., elevated liver enzyme concentrations, neutropenia, thrombocytopenia) occur in patients with GCA receiving IV or subcutaneous tocilizumab, interruption of tocilizumab therapy, dose reduction, or discontinuance of therapy may be required ((see Tables 1–3).1)

Cytokine Release Syndrome Associated with Chimeric Antigen Receptor-T cell Therapy

For the management of CAR T cell-induced severe or life-threatening CRS, tocilizumab should only be given IV as an infusion over 60 minutes.(1) Subcutaneous administration is not labeled by the FDA for the treatment of CRS.(1) Tocilizumab may be used alone or in combination with corticosteroids.(1)

In patients 2 years of age or older who weigh less than 30 kg, the manufacturer recommends a tocilizumab dosage of 12 mg/kg given by IV infusion over 60 minutes.(1) In adults and pediatric patients 2 years of age or older who weigh 30 kg or more, the manufacturer recommends a tocilizumab dosage of 8 mg/kg given by IV infusion over 60 minutes.(1)

If clinical improvement in the signs and symptoms of CRS does not occur following the first dose of tocilizumab, up to 3 additional doses may be administered.(1) The interval between consecutive doses should be at least 8 hours.(1) Doses exceeding 800 mg per infusion are not recommended.(1)

Systemic Sclerosis-Associated Interstitial Lung Disease

The recommended dosage of tocilizumab in adults with SSc-ILD is 162 mg subcutaneously every week. (1)

Tocilizumab is not FDA-labeled for IV administration in patients with SSc-ILD.(1) The use of the prefilled pen has not been evaluated in patients with SSc-ILD.(1)

Treatment Interruptions or Discontinuance for Toxicity

If certain dose-related laboratory changes (i.e., elevated liver enzyme concentrations, neutropenia, thrombocytopenia) occur in patients with SSc-ILD receiving IV or subcutaneous tocilizumab, interruption of tocilizumab therapy, dose reduction, or discontinuance of therapy may be required ((see Tables 1–3).1)

Coronavirus Disease 2019 (COVID-19)

The recommended dosage of tocilizumab in adult patients with COVID-19 is 8 mg/kg given as a single IV infusion over 60 minutes.(1) If clinical signs or symptoms worsen or do not improve after the first dose, one additional infusion may be administered at least 8 hours after the initial infusion.(1) Doses >800 mg per infusion are not recommended.(1)

The FDA EUA permits administration of tocilizumab for treatment of COVID-19 in hospitalized pediatric patients( (2–18 years of age weighing less than 30 kg) in a dosage of 12 mg/kg (maximum of 800 mg per infusion) given as a single 60-minute IV infusion; one additional IV infusion may be administered at least 8 hours after the first infusion if clinical signs or symptoms worsen or do not improve after the initial dose.70) In hospitalized pediatric patients 2–18 years of age weighing 30 kg or more, the FDA EUA permits administration of tocilizumab in a dosage of 8 mg/kg (maximum of 800 mg per infusion) given as a single 60-minute IV infusion; one additional IV infusion may be administered at least 8 hours after the first infusion if clinical signs or symptoms worsen or do not improve after the initial dose.(70)

Tocilizumab is not authorized by the EUA or FDA-label for subcutaneous administration for treatment of COVID-19.(70)

Dosage Modification for Serious Infections or Laboratory Abnormalities

If a serious infection, an opportunistic infection, or sepsis develops, tocilizumab should be discontinued until the infection is controlled.(1)

Table 1. Recommended Dosage Adjustment Based on Liver Enzyme Abnormalities in Adults with Rheumatoid Arthritis, SSc-ILD, or GCA

ALT or AST Value

Recommendation for RA and SSc-ILD

Recommendation for GCA

>1 to 3 times ULN

Modify dosage of concomitant DMARDs if appropriate(1)

Modify dosage of concomitant immunomodulatory agents if appropriate(1)

 

For persistent increases within this range in patients receiving IV tocilizumab, reduce dose of tocilizumab to 4 mg/kg or withhold therapy until ALT/AST values normalize(1)

For persistent increases within this range in patients receiving IV tocilizumab, withhold therapy until ALT/AST values normalize(1)

 

For persistent increases within this range in patients receiving sub-Q tocilizumab, reduce frequency of tocilizumab administration to every other week or withhold therapy until ALT/AST values normalize; resume tocilizumab at every other week and increase frequency to every week as clinically indicated(1)

For persistent increases within this range in patients receiving sub-Q tocilizumab, reduce frequency of tocilizumab administration to every other week or withhold therapy until ALT/AST values normalize; resume tocilizumab at every other week and increase frequency to every week as clinically indicated(1)

>3 to 5 times ULN (confirmed by repeat testing)

Interrupt tocilizumab therapy until ALT/AST <3 times ULN and follow recommendations for ALT/AST >1 to 3 times ULN(1)

Interrupt tocilizumab therapy until ALT/AST <3 times ULN and follow recommendations for ALT/AST >1 to 3 times ULN(1)

 

For persistent increases of >3 times ULN, discontinue tocilizumab(1)

For persistent increases of >3 times ULN, discontinue tocilizumab(1)

>5 times ULN

Discontinue tocilizumab(1)

Discontinue tocilizumab(1)

Table 2. Recommended Dosage Adjustment Based on Absolute Neutrophil Count (ANC) in Adults with Rheumatoid Arthritis, GCA, or SSc-ILD

ANC (cells/mm3)

Recommendation for RA and SSc-ILD

Recommendation for GCA

>1000

Maintain current dosage(1)

Maintain current dosage(1)

500–1000

Interrupt tocilizumab therapy(1)

Interrupt tocilizumab therapy(1)

 

When ANC >1000/mm3 in patients receiving IV tocilizumab, resume tocilizumab at a dose of 4 mg/kg and increase to 8 mg/kg as clinically indicated(1)

When ANC >1000/mm3 in patients receiving IV tocilizumab, resume tocilizumab at a dose of 6 mg/kg(1)

 

When ANC >1000/mm3 in patients receiving sub-Q tocilizumab, resume tocilizumab at every other week and increase frequency to every week as clinically indicated(1)

When ANC >1000/mm3 in patients receiving sub-Q tocilizumab, resume tocilizumab at every other week and increase frequency to every week as clinically indicated(1)

<500

Discontinue tocilizumab(1)

Discontinue tocilizumab(1)

Table 3. Recommended Dosage Adjustment Based on Platelet Count in Adults with Rheumatoid Arthritis, GCA, or SSc-ILD

Platelet Count (cells/mm3)

Recommendation for RA and SSc-ILD

Recommendation for GCA

50,000–100,000

Interrupt tocilizumab therapy(1)

Interrupt tocilizumab therapy(1)

 

When platelet count >100,000/mm3 in patients receiving IV tocilizumab, resume tocilizumab at a dose of 4 mg/kg and increase to 8 mg/kg as clinically indicated(1)

When platelet count >100,000/mm3 in patients receiving IV tocilizumab, resume tocilizumab at a dose of 6 mg/kg(1)

 

When platelet count >100,000/mm3 in patients receiving sub-Q tocilizumab, resume tocilizumab at every other week and increase frequency to every week as clinically appropriate(1)

When platelet count >100,000/mm3 in patients receiving sub-Q tocilizumab, resume tocilizumab at every other week and increase frequency to every week as clinically appropriate(1)

<50,000

Discontinue tocilizumab(1)

Discontinue tocilizumab(1)

Special Populations

Hepatic Impairment

Use of the drug in patients with active hepatic disease or hepatic impairment is not recommended.(1)

Renal Impairment

Dosage adjustment is not necessary in patients with mild or moderate renal impairment; use of tocilizumab in patients with severe renal impairment has not been evaluated.(1)

Geriatric Patients

The manufacturer makes no specific dosage recommendations for geriatric patients.(1)

Description

Tocilizumab, a recombinant humanized monoclonal antibody specific for interleukin-6 (IL-6) receptor, is a biologic response modifier and a disease-modifying antirheumatic drug (DMARD).(1)(3)(4)(5)(6)(9)(10)(11)(13) Tocilizumab is an IgG1 kappa immunoglobulin that binds to both soluble and membrane-bound IL-6 receptors and inhibits IL-6-mediated signaling through these receptors, thereby resulting in a reduction in inflammatory mediator production.(1)(3)(4)(5)(9)(10) The drug is produced in mammalian (Chinese hamster ovary) cells.(1)

IL-6, a pleiotropic proinflammatory cytokine, is produced by various cell types, including T-cells, B-lymphocytes, monocytes, fibroblasts, synoviocytes, and endothelial cells, and has a broad spectrum of biologic activities.(1)(3)(4)(5)(9)(10)(12) IL-6 is involved in T-cell activation, induction of immunoglobulin secretion, initiation of hepatic acute phase protein synthesis, stimulation of hematopoietic precursor cell proliferation and differentiation, and induction of osteoclast differentiation and activation.(1)(4)(5)(9)(10)(12) While the causes of rheumatoid arthritis have not been fully elucidated, proinflammatory cytokines, including IL-6, appear to play critical roles in the disease process.(4)(10)(12) IL-6 is overexpressed in synovial tissue in patients with rheumatoid arthritis and is thought to contribute to synovial proliferation and joint destruction in patients with the disease.(4)(9)(10)(12) Elevated levels of IL-6 in serum and synovial fluid have been shown to correlate with clinical and laboratory measures of disease activity in patients with rheumatoid arthritis.(3)(5)(9)(10)(12) IL-6 also is elevated in serum and synovial fluid in patients with polyarticular or systemic juvenile idiopathic arthritis (JIA), and the elevated levels of IL-6 have been correlated with disease activity.(20)(21)(26)(27)(28)

The pharmacokinetics of tocilizumab are characterized by nonlinear elimination that is a combination of linear clearance and Michaelis-Menten elimination.(1) The nonlinear part of the drug's elimination leads to an increase in drug exposure that is more than dose proportional.(1) The pharmacokinetics of tocilizumab do not change with time.(1) Because of the dependence of total clearance on tocilizumab serum concentrations, the half-life of the drug is also concentration dependent and varies depending on the serum concentration.(1) Population pharmacokinetic analyses in patient populations tested to date do not suggest any relationship between clearance of tocilizumab and the presence of anti-drug antibodies.(1) The apparent steady-state half-life of tocilizumab in adults with rheumatoid arthritis is up to 11 or up to 13 days following IV administration of tocilizumab 4 or 8 mg/kg, respectively, every 4 weeks and up to 13 or up to 5 days after subcutaneous administration of tocilizumab 162 mg every week or every other week, respectively.(1) In patients with giant cell arteritis (GCA), the effective half-life of tocilizumab at steady state was 18.3–18.9 days in patients receiving 162 mg subcutaneously once weekly and 4.2–7.9 days in those receiving 162 mg subcutaneously every other week.(1) For intravenous administration in GCA patients, the effective half-life of tocilizumab at steady state was 13.2 days in patients receiving 6 mg/kg every 4 weeks.(1) The reported half-life in pediatric patients with polyarticular or systemic juvenile idiopathic arthritis (JIA) following IV administration is up to 17 or up to 16 days, respectively, at steady state.(1) Population pharmacokinetic analyses in adult rheumatoid arthritis and GCA patients indicate that age, gender, and race do not affect the pharmacokinetics of tocilizumab.(1) However, linear clearance was found to increase with body size.(1) In rheumatoid arthritis patients, systemic drug exposure was substantially greater (86%) in patients weighing more than 100 kg than in those weighing less than 60 kg following administration of a weight-based IV dose of 8 mg/kg ((see Dosage and Administration: Dosage).1) At a fixed subcutaneous dosage, tocilizumab exposure is inversely related to body weight.(1)

Cautions

Contraindications

  • Tocilizumab is contraindicated in patients with known hypersensitivity to the drug.(1)

Warnings/Precautions

Warnings

Risk of Serious Infections

A boxed warning regarding the risk of serious infections is included in the prescribing information for tocilizumab.(1) Patients treated with tocilizumab are at an increased risk for developing serious infections that may lead to hospitalization or death.(1) Reported infections include active tuberculosis, invasive fungal infections including candidiasis, aspergillosis, and pneumocystis, bacterial, viral, and other infections due to opportunistic pathogens.(1)

The most common serious infections reported in patients receiving tocilizumab have included pneumonia, urinary tract infection, cellulitis, herpes zoster, gastroenteritis, diverticulitis, sepsis, and septic (bacterial) arthritis.(1) Opportunistic infections (e.g., tuberculosis, cryptococcus, aspergillosis, candidiasis, pneumocystosis) also have been reported in patients receiving tocilizumab.(1) Other serious infections may occur.(1) Patients have presented with disseminated rather than local disease; patients often were receiving concomitant therapy with immunosuppressive agents (e.g., methotrexate, corticosteroids) that, in addition to their underlying condition, could have predisposed them to infections.(1)

The risks and benefits of treatment with tocilizumab should be carefully considered prior to initiating therapy in patients with chronic or recurrent infection.(1) Patients should be closely monitored for the development of signs and symptoms of infection during and after treatment with tocilizumab, including the possible development of tuberculosis in patients who tested negative for latent tuberculosis infection prior to initiating therapy.(1)

Tocilizumab therapy should not be initiated in patients with active infections, including localized infections.(1) Clinicians should consider potential risks and benefits of the drug prior to initiating therapy in patients with a history of chronic, recurring, serious, or opportunistic infections; patients with underlying conditions that may predispose them to infections; and patients who have been exposed to tuberculosis or who reside or have traveled in regions where tuberculosis or mycoses are endemic.(1) Any patient who develops a new infection while receiving tocilizumab should undergo a thorough diagnostic evaluation (appropriate for an immunocompromised patient), appropriate anti-infective therapy should be initiated, and the patient should be closely monitored.(1) If a serious infection, an opportunistic infection, or sepsis develops, tocilizumab should be discontinued until the infection is controlled.(1) There is limited information regarding the use of tocilizumab in patients with COVID-19 and concomitant active serious infections; consider the risks and benefits of treatment with tocilizumab in COVID-19 patients with other concurrent infections (1)

Patients, except for those with COVID-19, should be evaluated for latent tuberculosis and for the presence of risk factors for tuberculosis prior to and periodically during therapy with tocilizumab.(1) When indicated, an appropriate antimycobacterial regimen for the treatment of latent tuberculosis infection should be initiated prior to tocilizumab therapy.(1) Antimycobacterial therapy should be considered prior to initiation of tocilizumab in individuals with a history of latent or active tuberculosis in whom an adequate course of antimycobacterial treatment cannot be confirmed and in individuals with a negative test for latent tuberculosis who have risk factors for tuberculosis.(1) Consultation with a tuberculosis specialist is recommended when deciding whether antimicrobial therapy should be initiated.(1) Patients receiving tocilizumab, including individuals with a negative test for latent tuberculosis, should be monitored for signs and symptoms of active tuberculosis.(1)

Viral reactivation can occur in patients receiving immunosuppressive therapies.(1) Herpes zoster exacerbation has been reported in patients receiving tocilizumab.(1) No cases of hepatitis B reactivation were observed in clinical trials of tocilizumab; however, patients who screened positive for hepatitis were excluded from these studies.(1)

Other Warnings and Precautions

GI Perforation

GI perforation has been reported in patients receiving tocilizumab in clinical trials, usually as a complication of diverticulitis.(1) Most patients who experienced GI perforation were receiving concomitant therapy with nonsteroidal anti-inflammatory agents (NSAIAs), corticosteroids, or methotrexate.(1) The relative contribution of these agents versus IV tocilizumab to the occurrence of GI perforation remains to be determined.(1)

Caution is advised if tocilizumab is used in patients at risk for GI perforation.(1) Patients who experience new-onset abdominal symptoms should be promptly evaluated for GI perforation.(1)

Hepatotoxicity

Serious hepatic injury has been observed in patients receiving IV or subcutaneous tocilizumab.(1) Some of these cases have resulted in a liver transplantation or death.(1) The time to onset for these cases ranged from months to years after initiation of tocilizumab therapy.(1) While most cases presented with marked elevations of aminotransferase concentrations (exceeding 5 times the upper limit of normal [ULN]), some cases presented with signs or symptoms of hepatic dysfunction and only mildly elevated aminotransferases.(1) In randomized controlled studies, tocilizumab treatment was associated with a higher incidence of aminotransferase elevations compared with placebo.(1) The incidence and magnitude of aminotransferase elevations were increased when tocilizumab was used in conjunction with a potentially hepatotoxic drug (e.g., methotrexate).(1)

In adults with rheumatoid arthritis, giant cell arteritis (GCA), or systemic sclerosis-associated interstitial lung disease (SSc-ILD), perform liver function tests (i.e., serum ALT and AST, alkaline phosphatase, and total bilirubin concentrations) before initiating tocilizumab therapy and then monitor every 4–8 weeks after initiation of therapy for the first 6 months of treatment and every 3 months thereafter.(1) The manufacturer recommends that tocilizumab therapy not be initiated in rheumatoid arthritis, GCA, or SSc-ILD patients with elevated aminotransferase concentrations greater than 1.5 times the ULN.(1) In patients who develop elevated ALT or AST concentrations exceeding 5 times the ULN, tocilizumab should be discontinued.(1) A similar pattern of serum aminotransferase elevations has been observed with tocilizumab therapy in patients with polyarticular or systemic JIA.(1) In such patients, liver function tests should be monitored at the time of the second infusion and every 4–8 weeks thereafter in patients with polyarticular JIA and every 2–4 weeks thereafter in patients with systemic JIA.(1)

In patients hospitalized with COVID-19, perform liver function tests (i.e., serum ALT and AST) before initiating tocilizumab therapy and during treatment.(1) The manufacturer recommends tocilizumab therapy should not be initiated in COVID-19 patients with elevated ALT or AST above 10 times the ULN.(1) Multi-organ failure with involvement of the liver is recognized as a complication of severe COVID-19.(1) The decision to administer tocilizumab should balance the potential benefit of treating COVID-19 against the potential risks of acute treatment with tocilizumab.(1)

In patients with symptoms indicative of possible liver injury (e.g., fatigue, anorexia, right upper abdominal discomfort, dark urine, jaundice), liver function tests should be performed promptly.(1) If the patient is found to have abnormal liver function tests (e.g., ALT greater than 3 times the ULN, total bilirubin greater than 2 times the ULN), tocilizumab therapy should be interrupted and the patient evaluated to determine the probable cause.(1) Therapy should only be restarted in patients with another explanation for their liver function test abnormalities after normalization of the liver function test results.(1)

Hematologic Effects

Treatment with tocilizumab has been associated with a higher incidence of neutropenia.(1) Reduction in neutrophil count to less than 1000/mm3 has been reported during clinical trials in patients receiving tocilizumab.(1) Infections have been infrequently reported in association with treatment-related neutropenia in long-term extension studies and postmarketing clinical experience.(1)

Reductions in platelet counts also have been reported in patients receiving tocilizumab.(1) In clinical trials of the drug, decreases in platelet counts were not associated with severe bleeding.(1)

Tocilizumab should not be initiated in RA, GCA, and SSc-ILD patients with an absolute neutrophil count less than 2000/mm3.(1) Treatment should be discontinued in patients who developed an absolute neutrophil count less than 500/mm3.(1)

Neutrophil and platelet counts should be monitored 4–8 weeks after initiation of tocilizumab therapy and every 3 months thereafter in adults with rheumatoid arthritis, GCA, or SSc-ILD.(1) Monitor neutrophils and platelet counts at the time of the second administration and every 4–8 weeks thereafter in patients with PJIA and every 2–4 weeks thereafter in patients with SJIA.(1) In patients with neutropenia or thrombocytopenia, dosage adjustment, treatment interruption, or discontinuance of the drug may be necessary.(1)

It is not recommended to initiate tocilizumab treatment in COVID-19 patients with an absolute neutrophil count less than 1000/mm3 or a platelet count less than 50,000/mm3.(1) Neutrophil and platelet counts should be monitored.(1)

Effects on Serum Lipids

Increased serum concentrations of total cholesterol, triglycerides, low-density lipoprotein (LDL)-cholesterol, and/or high-density lipoprotein (HDL)-cholesterol have been reported in patients receiving tocilizumab.(1)

Lipoprotein concentrations should be monitored approximately 4–8 weeks after initiation of tocilizumab therapy.(1) Lipid disorders should be managed as clinically appropriate.(1)

Because tocilizumab may adversely affect lipid profiles and also elevate blood pressure in some patients, a postmarketing, randomized, open-label, multicenter cardiovascular outcomes study was conducted in patients with moderate to severe rheumatoid arthritis.(1)(62) This cardiovascular safety study was designed to exclude a moderate increase in cardiovascular risk in patients treated with tocilizumab compared with the TNF blocking agent etanercept.(1) The study included 3,080 seropositive rheumatoid arthritis patients with active disease and an inadequate response to nonbiologic DMARDs who were 50 years of age or older with at least one additional cardiovascular risk factor beyond rheumatoid arthritis.(1) Patients were randomized in a 1:1 ratio to either IV tocilizumab 8 mg/kg every 4 weeks or subcutaneous etanercept 50 mg every week and then followed for an average of 3.2 years.(1) The primary end point was the comparison of the time-to-first occurrence of any component of a composite of major adverse cardiovascular events (e.g., nonfatal myocardial infarction, nonfatal stroke, cardiovascular death); the final intent-to-treat analysis was based on a total of 161 confirmed cardiovascular events (83 [5.4%] for tocilizumab and 78 [5.1%] for etanercept) reviewed by an independent and blinded adjudication committee.(1) Non-inferiority of tocilizumab to etanercept for cardiovascular risk was determined by excluding a more than 80% relative increase in the risk of major adverse cardiovascular events.(1) The estimated hazard ratio for this risk comparing tocilizumab to etanercept was 1.05 (95% confidence interval [0.77, 1.43]).(1)

Malignancies

Immunosuppressive therapy may increase the risk of malignancies.(1) Whether treatment with tocilizumab affects development of malignancies remains to be determined.(1) Malignancies were reported in clinical trials of the drug.(1)

Sensitivity Reactions

Hypersensitivity reactions, including anaphylaxis, have been reported in patients receiving tocilizumab; fatal anaphylactic reactions have occurred in those receiving IV infusions of the drug.(1)(15) During clinical trials of tocilizumab, hypersensitivity reactions requiring treatment discontinuance (e.g., anaphylaxis, generalized erythema, rash, urticaria) have been reported in various patient populations and with either IV or subcutaneous therapy.(1) During postmarketing experience, hypersensitivity reactions, including anaphylaxis and death, have occurred in patients receiving various IV dosages, either with or without concomitant therapy, including in patients who received premedication.(1) Hypersensitivity reactions, including anaphylaxis, have occurred both with and without prior hypersensitivity reactions and as early as the first IV infusion of the drug.(1)

Appropriate agents and equipment should be available for immediate use in case a serious hypersensitivity reaction occurs during IV infusion of the drug.(1) Patients receiving subcutaneous therapy with tocilizumab should be advised to seek immediate medical attention if they experience symptoms of a hypersensitivity reaction.(1) If a hypersensitivity reaction occurs, administration of the drug should be stopped immediately and the drug should be permanently discontinued.(1) Tocilizumab is contraindicated in patients with known hypersensitivity to the drug.(1)

Demyelinating Disorders

The effect of tocilizumab on demyelinating disorders remains to be determined.(1) Multiple sclerosis and chronic inflammatory demyelinating polyneuropathy have been reported rarely in patients with rheumatoid arthritis receiving tocilizumab in clinical trials.(1) Patients receiving tocilizumab should be monitored for signs and symptoms suggestive of a demyelinating disorder.(1) Clinicians should exercise caution when considering tocilizumab therapy in patients with preexisting or recent-onset demyelinating disorders.(1)

Immunization

Live vaccines should be avoided in patients receiving tocilizumab.(1) Vaccinations should be brought up to date prior to initiation of the drug.(1)

Immunogenicity

In clinical trials evaluating IV tocilizumab in patients with rheumatoid arthritis, antibodies to the drug were detected in about 2% of tocilizumab-treated patients; about 11% of these patients experienced hypersensitivity reactions resulting in drug discontinuance.(1) Neutralizing antibodies developed in about 1% of such patients receiving the drug IV.(1) In clinical trials evaluating subcutaneous tocilizumab in patients with rheumatoid arthritis, antibodies to the drug were detected in 0.9% of patients receiving the drug; neutralizing antibodies developed in about 0.8% of patients receiving subcutaneous tocilizumab.(1) No relationship between antibody development and adverse effects or loss of clinical response was observed, and detection rates were similar with IV or subcutaneous therapy.(1)

In clinical trials evaluating subcutaneous tocilizumab in patients with SSc-ILD, antibodies to tocilizumab were detected in about 1.8% of patients.(1) Antibodies were neutralizing, and none of the patients experienced hypersensitivity reactions.(1)

In clinical trials evaluating IV tocilizumab in patients with polyarticular JIA, one patient developed antibodies to the drug without developing a hypersensitivity reaction.(1) In clinical trials evaluating subcutaneous tocilizumab in patients with polyarticular JIA, 3 patients developed neutralizing antibodies without developing a serious or clinically important hypersensitivity reaction.(1) In clinical trials evaluating IV tocilizumab in patients with systemic JIA, 2 patients (2%) developed neutralizing antibodies, with urticaria and angioedema occurring in one of the patients and macrophage activation syndrome while on escape therapy reported in the other patient.(1)

Specific Populations

Pregnancy

A pregnancy exposure registry has been established by the manufacturer that monitors pregnancy outcomes in women receiving tocilizumab.(1) Clinicians or pregnant patients can register at 877-311-8972.(1)

Limited data regarding tocilizumab use in pregnant women are not sufficient to determine whether there is a drug-associated risk for major birth defects and miscarriage.(1) Monoclonal antibodies such as tocilizumab are actively transported across the placenta during the third trimester of pregnancy and may affect immune response in infants exposed to the drugs in utero.(1)

In animal reproduction studies, IV administration of tocilizumab to cynomolgus monkeys during organogenesis caused abortion/embryofetal death at dosages of 1.25 (or more) times the maximum recommended human IV dosage of 8 mg/kg every 2–4 weeks.(1) Evidence in animals suggests that inhibition of interleukin-6 (IL-6) signaling may interfere with cervical ripening and dilatation and myometrial contractile activity, which could potentially result in parturition delays.(1) Based on animal data, there may be a potential risk to the fetus.(1)

As pregnancy progresses, monoclonal antibodies are increasingly transported across the placenta, with the largest amount transferred during the third trimester.(1) The risks and benefits of administering live or live attenuated vaccines to infants exposed to tocilizumab in utero should be considered, since the safety of such vaccines in infants exposed to the drug in utero is unknown.(1)

Lactation

It is not known whether tocilizumab is distributed into milk, and the effects of the drug on the breast-fed infant and on milk production also are not known.(1) Maternal immunoglobulin G (IgG) is distributed into human milk.(1) If tocilizumab is distributed into human milk, the effects of local exposure in the GI tract and potential limited systemic exposure in the infant are unknown.(1) The lack of clinical data during lactation precludes a clear determination of the risk of tocilizumab exposure to nursing infants.(1) Therefore, the manufacturer states that the developmental and health benefits of breast-feeding should be considered along with the mother's clinical need for tocilizumab and any potential adverse effects on the breast-fed child from the drug or from the underlying maternal condition.(1)

Pediatric Use

Safety and efficacy of IV and subcutaneous tocilizumab for the management of active polyarticular or systemic JIA and safety and efficacy of IV tocilizumab for the management of severe or life-threatening chimeric antigen receptor (CAR) T cell-induced cytokine release syndrome (CRS) have been established in pediatric patients 2 years of age and older.(1)(20)(21) Safety and efficacy of the drug in children younger than 2 years of age or for the management of conditions other than polyarticular or systemic JIA and CAR T cell-induced CRS in children have not been established.(1)

A multicenter, open-label, single-arm study to evaluate the pharmacokinetics, safety, pharmacodynamics, and efficacy of tocilizumab over 12 weeks in 11 patients with systemic JIA under 2 years of age was conducted.(1) Patients received IV tocilizumab 12 mg/kg every 2 weeks, and concurrent use of stable background treatment with corticosteroids, methotrexate, and/or nonsteroidal anti-inflammatory agents was permitted.(1) Patients who completed the 12-week phase of the study could continue to an optional extension phase for a total of 52 weeks or until the age of 2 years, whichever was longer.(1) The primary pharmacokinetic parameters (peak and trough concentrations and AUC) of tocilizumab at steady state in these patients under 2 years of age were within the ranges of these parameters observed in patients 2–17 years of age with systemic JIA.(1) In patients under 2 years of age with systemic JIA, serious adverse effects, adverse effects leading to drug discontinuance, and infectious adverse effects were reported in 27.3, 36.4, and 81.8% of patients, respectively.(1) Six patients (54.5%) experienced hypersensitivity reactions and 3 of these patients experienced serious hypersensitivity reactions and were withdrawn from the study.(1) Three of the patients who developed hypersensitivity reactions (two with serious hypersensitivity reactions) developed treatment-induced anti-tocilizumab antibodies after the event.(1) There were no cases of macrophage activation syndrome (MAS) based on the study protocol-specified criteria, but there were 2 cases of suspected MAS based on Ravelli criteria.(1)

In a retrospective analysis of pooled outcome data for patients treated with IV tocilizumab for CAR T cell-induced CRS, 25 patients were children (2–12 years of age) and 17 patients were adolescents (12–18 years of age).(1) There were no differences in safety or efficacy between the pediatric patients and the adults.(1)

Geriatric Use

Approximately 16% of patients with rheumatoid arthritis who received IV tocilizumab in clinical trials were ≥65 years of age and about 2% were ≥75 years of age.(1) Approximately 28% of patients with rheumatoid arthritis who received subcutaneous tocilizumab in clinical trials were ≥65 years of age and about 4% were ≥75 years of age.(1) Serious infection occurred with greater frequency in those ≥65 years of age relative to younger adults.(1) Because the geriatric population in general may have a higher incidence of infections than younger adults, tocilizumab should be used with caution in this age group.(1)

Clinical studies that evaluated tocilizumab for CAR T cell-induced CRS did not include sufficient numbers of patients ≥65 years of age to determine whether they respond differently than younger adults.(1)

Population pharmacokinetic analyses indicate that age does not affect the pharmacokinetics of tocilizumab.(1)

Hepatic Impairment

Safety and efficacy of tocilizumab have not been established in patients with hepatic impairment, including those with serologic evidence of hepatitis B virus (HBV) or hepatitis C virus (HCV) infection.(1) Use of the drug in patients with active hepatic disease or hepatic impairment is not recommended.(1)

The pharmacokinetics of tocilizumab have not been formally studied in patients with hepatic impairment.(1)

Renal Impairment

The pharmacokinetics of tocilizumab have not been formally studied in patients with renal impairment.(1) Most of the patients with rheumatoid arthritis, GCA, and SSc-ILD in the population pharmacokinetic analysis had normal renal function or mild renal impairment.(1) Mild renal impairment (creatinine clearance less than 80 mL/minute but not less than 50 mL/minute) did not appear to affect the pharmacokinetics of the drug.(1) About one-third of the patients in the GCA trial had moderate renal impairment at baseline (creatinine clearance 30–59 mL/minute); no effect on tocilizumab exposure was noted in these patients.(1)

No dosage adjustment is required in patients with mild or moderate renal impairment.(1) Tocilizumab has not been evaluated in patients with severe renal impairment to date.(1)

Common Adverse Effects

Adverse effects reported in 5% or more of patients receiving tocilizumab include upper respiratory tract infection,(1) nasopharyngitis,(1) headache,(1) hypertension,(1) increased ALT concentrations,(1) and injection site reactions (e.g., erythema, pruritus, pain, hematoma).(1)

Drug Interactions

Drugs Metabolized by Hepatic Microsomal Enzymes

Possible increased metabolism of drugs that are metabolized by cytochrome P-450 (CYP) isoenzymes.(1) Because cytokines such as interleukin-6 (IL-6) may down-regulate CYP enzymes, inhibition of IL-6 by tocilizumab in patients with rheumatoid arthritis may restore CYP enzyme activity to higher levels.(1) In vitro studies indicate that tocilizumab may alter expression of CYP isoenzymes including 1A2, 2B6, 2C9, 2C19, 2D6, and 3A4; effects of the drug on CYP2C8 or transporters (e.g., P-glycoprotein) have not been elucidated.(1) Effects of tocilizumab on CYP enzyme activity may persist for several weeks after the drug is discontinued.(1)

Following initiation or discontinuance of tocilizumab therapy, patients receiving certain drugs metabolized by CYP isoenzymes (i.e., those with a low therapeutic index that require individualized dosing [e.g., cyclosporine, theophylline, warfarin]) should be monitored for therapeutic effect and/or changes in serum concentrations, and dosages of these drugs should be adjusted as needed.(1) Caution also is advised when tocilizumab is used concomitantly with CYP3A4 substrates (e.g., oral contraceptives, atorvastatin, lovastatin) for which a reduction in efficacy would be undesirable.(1)

Dextromethorphan

Pharmacokinetic interaction (possible decreased exposure to dextromethorphan and/or dextrorphan).(1) In patients with rheumatoid arthritis who are not receiving tocilizumab, administration of dextromethorphan 30 mg results in systemic exposure to dextromethorphan (a substrate of CYP2D6 and CYP3A4) that is similar to that observed in healthy individuals; however, exposure to the dextrorphan metabolite (a substrate of CYP3A4) is substantially reduced in rheumatoid arthritis patients.(1) One week following IV administration of a single 8-mg/kg dose of tocilizumab, exposures to dextromethorphan and dextrorphan were decreased by 5 and 29%, respectively.(1)

Omeprazole

Pharmacokinetic interaction (possible decreased exposure to omeprazole following initiation of tocilizumab therapy).(1) In patients with rheumatoid arthritis who are not receiving tocilizumab, administration of omeprazole 10 mg results in systemic exposure to omeprazole (a substrate of CYP2C19 and CYP3A4) that is approximately twofold higher than values observed in healthy individuals.(1) In patients with rheumatoid arthritis who received 10 mg of omeprazole before and one week after IV administration of an 8-mg/kg dose of tocilizumab, systemic exposure to omeprazole (as measured by area under the plasma concentration-time curve [AUC]) decreased by 12% in poor and intermediate metabolizers of the drug and by 28% in extensive metabolizers; AUC values for omeprazole observed following the tocilizumab dose were slightly higher than values observed after omeprazole administration in healthy individuals.(1)

Simvastatin

Pharmacokinetic interaction (possible decreased exposure to simvastatin and simvastatin acid following initiation of tocilizumab therapy).(1) In patients with rheumatoid arthritis who are not receiving tocilizumab, administration of simvastatin 40 mg results in systemic exposures to simvastatin (a substrate of CYP3A4 and organic anion transporter [OATP] 1B1) and its metabolite, simvastatin acid, that are approximately fourfold to tenfold higher and twofold higher, respectively, than values observed in healthy individuals.(1) One week after rheumatoid arthritis patients received a single 10-mg/kg IV dose of tocilizumab, systemic exposures to simvastatin and simvastatin acid decreased by 57 and 39%, respectively, and values in the tocilizumab-treated patients were similar to or slightly higher than values observed after simvastatin administration in healthy individuals.(1) Following discontinuance of tocilizumab in rheumatoid arthritis patients, systemic exposure to simvastatin and simvastatin acid increased.(1) When selecting simvastatin dosages for patients with rheumatoid arthritis, clinicians should consider the potential for altered systemic exposure to the drug following initiation or discontinuance of tocilizumab therapy.(1)

Antirheumatic Drugs

In rheumatoid arthritis patients, population pharmacokinetic analyses indicated that concomitant use of methotrexate, corticosteroids, or nonsteroidal anti-inflammatory agents (NSAIAs) does not appear to affect clearance of tocilizumab.(1) Concomitant use of methotrexate (10–25 mg once weekly) and tocilizumab (single 10-mg/kg IV dose) did not substantially affect exposure to methotrexate.(1) An increased incidence and magnitude of serum aminotransferase elevations were observed when potentially hepatotoxic drugs such as methotrexate were used in combination with tocilizumab.(1)

Tocilizumab has not been studied in conjunction with biologic disease-modifying antirheumatic drugs (DMARDs), including tumor necrosis factor (TNF; TNF-α) blocking agents, interleukin-1 (IL-1) receptor antagonists, anti-CD20 monoclonal antibodies, and selective costimulation modulators.(1) Because of the possibility of increased immunosuppression and an increased risk of infection, concomitant use of tocilizumab with biologic DMARDs should be avoided.(1)

Vaccines

Live vaccines should not be administered to patients receiving tocilizumab.(1) Because inhibition of interleukin-6 (IL-6) may interfere with the normal immune response to new antigens, all patients (particularly pediatric and geriatric patients) should receive all appropriate vaccines recommended by current immunization guidelines prior to initiation of tocilizumab therapy, if possible.(1) The interval between administration of live vaccines and initiation of tocilizumab therapy should be in accordance with current vaccination guidelines regarding immunosuppressive agents.(1) Information is not available regarding immune response to vaccines in patients receiving tocilizumab, nor is information available regarding secondary transmission of infection from individuals receiving live vaccines to patients receiving tocilizumab.(1)

Additional Information

The American Society of Health-System Pharmacists, Inc. represents that the information provided in the accompanying monograph was formulated with a reasonable standard of care, and in conformity with professional standards in the field. Readers are advised that decisions regarding use of drugs are complex medical decisions requiring the independent, informed decision of an appropriate health care professional, and that the information contained in the monograph is provided for informational purposes only. The manufacturer’s labeling should be consulted for more detailed information. The American Society of Health-System Pharmacists, Inc. does not endorse or recommend the use of any drug. The information contained in the monograph is not a substitute for medical care.

Preparations

Excipients in commercially available drug preparations may have clinically important effects in some individuals; consult specific product labeling for details.

Please refer to the ASHP Drug Shortages Resource Center for information on shortages of one or more of these preparations.

Tocilizumab is obtained through designated specialty pharmacies.(88) Contact manufacturer for specific availability information.(88)

Tocilizumab (recombinant)

Routes

Dosage Forms

Strengths

Brand Names

Manufacturer

Parenteral

Injection, for subcutaneous use

162 mg/0.9 mL

Actemra® (available as single-use prefilled syringe)

Genentech

162 mg/0.9 mL

Actemra® ACTPen® (available as single-use prefilled autoinjector)

Genentech

Injection concentrate, for IV infusion

20 mg/mL (80 mg, 200 mg, 400 mg)

Actemra®

Genentech