Truxima

Truxima

rituximab

Manufacturer:

Celltrion Healthcare

Distributor:

Zuellig Pharma
Full Prescribing Info
Contents
Rituximab.
Description
Active ingredient: rituximab.
Truxima is supplied in sterile, preservative-free, non-pyrogenic single-dose vial.
Single-dose vials: Vials contain 100 mg/10 ml and 500 mg/50 ml.
Sterile/Radioactive Statement: Sterile.
Excipients/Inactive Ingredients: Sodium citrate, polysorbate 80, sodium chloride, sodium hydroxide, hydrochloric acid, water for injections.
Action
Therapeutic/Pharmacologic Class of Drug: Antineoplastic agents, monoclonal antibodies. ATC Code: L01FA01.
PHARMACOLOGY: Pharmacodynamics: Mechanism of Action: Rituximab is a chimeric mouse/human monoclonal antibody that binds specifically to the transmembrane antigen CD20. This antigen is located on pre-B- and mature B-lymphocytes, but not on haemopoietic stem cells, pro-B-cells, normal plasma cells or other normal tissue. The antigen is expressed on >95% of all B-cell non-Hodgkin's lymphomas (NHLs). Following antibody binding, CD20 is not internalized or shed from the cell membrane into the environment. CD20 does not circulate in the plasma as a free antigen and, thus, does not compete for antibody binding.
Rituximab binds to the CD20 antigen on B-lymphocytes and initiates immunologic reactions that mediate B-cell lysis. Possible mechanisms of cell lysis include complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), and induction of apoptosis. Finally, in vitro studies have demonstrated that rituximab sensitizes drug-resistant human B-cell lymphoma lines to the cytotoxic effects of some chemotherapeutic agents.
Peripheral B-cell counts declined to levels below normal following the first dose of rituximab. In patients treated for haematological malignancies, B-cell recovery began within 6 months of treatment returning to normal levels within 6 months of treatment and generally returning to normal levels, within 12 months after completion of therapy, although in some patients this may take longer (see Clinical Trials: Experience from Clinical Trials in Haemato-Oncology under Adverse Reactions).
In patients with rheumatoid arthritis, the duration of peripheral B cell depletion was variable. The majority of patients received further treatment prior to full B cell repletion. A small proportion of patients had prolonged peripheral B-cell depletion lasting 2 years or more after their last dose of rituximab.
In GPA and MPA patients, peripheral blood CD19 B-cells depleted to less than 10 cells/µl following the first two infusions of rituximab and remained at that level in most patients through month 6.
Of 67 patients evaluated for human anti-mouse antibody (HAMA), none were positive. Of 356 non-Hodgkin's lymphoma patients evaluated for human anti-chimeric antibody (HACA) 1.1% (4 patients) were positive.
Clinical/Efficacy Studies: Low-grade or follicular non-Hodgkin's lymphoma: Monotherapy: Initial treatment, weekly for 4 doses: In the pivotal study, 166 patients with relapsed or chemoresistant low-grade or follicular B-cell NHL received 375 mg/m2 of rituximab as an IV infusion weekly for four doses. The overall response rate (ORR) in the intent-to-treat (ITT) population was 48% (CI95% 41% - 56%) with a 6% complete response (CR) and a 42% partial response (PR) rate. The projected median time to progression (TTP) for responding patients was 13.0 months.
In a subgroup analysis, the ORR was higher in patients with IWF B, C, and D histologic subtypes as compared to IWF A subtype (58% versus 12%), higher in patients whose largest lesion was < 5 cm versus > 7 cm in greatest diameter (53% versus 38%), and higher in patients with chemosensitive relapse as compared to chemoresistant (defined as duration of response < 3 months) relapse (50% versus 22%). ORR in patients previously treated with autologous bone marrow transplant (ABMT) was 78% versus 43% in patients with no ABMT. Neither age, sex, lymphoma grade, initial diagnosis, presence nor absence of bulky disease, normal or high LDH nor presence of extranodal disease had a statistically significant effect (Fisher's exact test) on response to rituximab. A statistically significant correlation was noted between response rates and bone marrow involvement. Forty percent of patients with bone marrow involvement responded compared to 59% of patients with no bone marrow involvement (p = 0.0186). This finding was not supported by a stepwise logistic regression analysis in which the following factors were identified as prognostic factors: histologic type, bcl-2 positivity at baseline, resistance to last chemotherapy and bulky disease.
Initial treatment, weekly for 8 doses: In a multi-center, single-arm study, 37 patients with relapsed or chemoresistant, low grade or follicular B-cell NHL received 375 mg/m2 of rituximab as IV infusion weekly for eight doses. The ORR was 57% (CI95% 41% - 73%; CR 14%, PR 43%) with a projected median TTP for responding patients of 19.4 months (range 5.3 to 38.9 months).
Initial treatment, bulky disease, weekly for 4 doses: In pooled data from three studies, 39 patients with relapsed or chemoresistant, bulky disease (single lesion ≥ 10 cm in diameter), low grade or follicular B-cell NHL received 375 mg/m2 of rituximab as IV infusion weekly for four doses. The ORR was 36% (CI95% 21% - 51%; CR 3%, PR 33%) with a median TTP for responding patients of 9.6 months (range 4.5 to 26.8 months).
Re-treatment, weekly for 4 doses: In a multicenter, single-arm study, 58 patients with relapsed or chemoresistant low grade or follicular B-cell NHL, who had achieved an objective clinical response to a prior course of rituximab, were re-treated with 375 mg/m2 of rituximab as IV infusion weekly for four doses. Three of the patients had received two courses of rituximab before enrollment and thus were given a third course in the study. Two patients were re-treated twice in the study. For the 60 re-treatments on study, the ORR was 38% (CI95% 26% - 51%; 10% CR, 28% PR) with a projected median TTP for responding patients of 17.8 months (range 5.4 - 26.6). This compares favorably with the TTP achieved after the prior course of rituximab (12.4 months).
Rituximab in combination with chemotherapy: Initial treatment: In an open-label randomized trial, a total of 322 previously untreated patients with follicular lymphoma were randomized to receive either CVP chemotherapy (cyclophosphamide 750 mg/m2, vincristine 1.4 mg/m2 up to a maximum of 2 mg on day 1, and prednisolone 40 mg/m2/day on days 1 - 5) every 3 weeks for 8 cycles or rituximab 375 mg/m2 in combination with CVP (R-CVP). Rituximab was administered on the first day of each treatment cycle. A total of 321 patients (162 R-CVP, 159 CVP) received therapy and were analyzed for efficacy. The median follow-up of patients was 53 months, R-CVP led to a significant benefit over CVP for the primary endpoint, time to treatment failure (27 months versus 6.6 months, p < 0.0001, log-rank test). The proportion of patients with a tumour response (CR, CRu, PR) was significantly higher (p < 0.0001, Chi-Square test) in the R-CVP group (80.9%) than the CVP group (57.2%). Treatment with R-CVP significantly prolonged the time to disease progression or death compared to CVP, 33.6 months and 14.7 months, respectively (p < 0.0001, log-rank test). The median duration of response was 37.7 months in the R-CVP group and was 13.5 months in the CVP group (p < 0.0001, log-rank test). The difference between the treatment groups with respect to overall survival showed a strong clinical benefit (p = 0.029, log-rank test stratified by center): survival rates at 53 months were 80.9% for patients in the R-CVP group compared to 71.1% for patients in the CVP group.
Results from three other randomized trials using rituximab in combination with chemotherapy regimen other than CVP (CHOP, MCP, CHVP/Interferon α) have also demonstrated significant improvements in response rates, time-dependent parameters as well as in overall survival. Key results from all four studies are summarized in Table 1 as follows. (See Table 1.)

Click on icon to see table/diagram/image

Maintenance therapy: Previously untreated follicular NHL: In a prospective, open label, international, multicenter, phase III trial 1193 patients previously untreated advanced follicular lymphoma received induction therapy with R-CHOP (n = 881), R-CVP (n = 268) or R-FCM (n = 44), according to the investigators' choice. A total of 1078 patients responded to induction therapy, of which 1018 were randomized to rituximab maintenance therapy (n = 505) or observation (n = 513). The two treatment groups were well balanced with regards to baseline characteristics and disease status. Rituximab maintenance treatment consisted of a single infusion of rituximab at 375 mg/m2 body surface area given every 2 months until disease progression or for a maximum period of two years.
After a median observation time of 25 months from randomization, maintenance therapy with rituximab resulted in a clinically relevant and statistically significant improvement in the primary endpoint of investigator assessed progression-free survival (PFS) as compared to no maintenance therapy in patients with previously untreated follicular NHL. This improvement in PFS was confirmed by an independent review committee (IRC) (Table 2). Significant benefit from maintenance treatment with rituximab was also seen for the secondary endpoints event-free survival (EFS), time to next anti-lymphoma treatment (TNLT) time to next chemotherapy (TNCT) and overall response rate (ORR) (Table 2).
The updated analysis corresponding to a median observation time of 73 months from randomization confirm the results of the primary analysis (Table 2). (See Table 2.)

Click on icon to see table/diagram/image

Rituximab maintenance treatment provided consistent benefit in all subgroups tested: gender (male, female), age (<60 years, ≥60 years), FLIPI score (1, 2 or 3), induction therapy (R-CHOP, R-CVP or R-FCM) and regardless of the quality of response to induction treatment (CR or PR).
Relapsed/Refractory follicular NHL: In a prospective, open label, international, multicentre, phase III trial, 465 patients with relapsed/refractory follicular NHL were randomised. In a first step to induction therapy with either CHOP (cyclophosphamide, doxorubicin, vincristine, prednisolone; n = 231) or rituximab plus CHOP (R-CHOP, n = 234). The two treatment groups were well balanced with regard to baseline characteristics and disease status. A total of 334 patients achieving a complete or partial remission following induction therapy were randomised in a second step to rituximab maintenance therapy (n=167) or observation (n=167). Rituximab maintenance treatment consisted of a single infusion of rituximab at 375 mg/m2 body surface area given every 3 months until disease progression or for a maximum period of two years.
The final efficacy analysis included all patients randomized to both parts of the study. After a median observation time of 31 months for patients randomised to the induction phase, R-CHOP significantly improved the outcome of patients with relapsed/refractory follicular NHL when compared to CHOP (Table 3). (See Table 3.)

Click on icon to see table/diagram/image

For patients randomized to the maintenance phase of the trial, the median observation time was 28 months from maintenance randomisation. Maintenance treatment with rituximab led to a clinically relevant and statistically significant improvement in the primary endpoint, PFS, (time from maintenance randomisation to relapse, disease progression or death) when compared to observation alone (p < 0.0001, log-rank test). The median PFS was 42.2 months in the rituximab maintenance arm compared to 14.3 months in the observation arm. Using a cox regression analysis, the risk of experiencing progressive disease or death was reduced by 61% with rituximab maintenance treatment when compared to observation (95% CI; 45% - 72%).
Kaplan-Meier estimated progression-free rates at 12 months were 78% in the rituximab maintenance group versus 57% in the observation group. An analysis of overall survival confirmed the significant benefit of rituximab maintenance over observation (p = 0.0039, log-rank test). Rituximab maintenance treatment reduced the risk of death by 56% (95% CI; 22%-75%).
The median time to new anti-lymphoma treatment was significantly longer with rituximab maintenance treatment than with observation (38.8 months versus 20.1 months, p < 0.0001, log-rank test). The risk of starting a new treatment was reduced by 50% (95% CI; 30% - 64%). In patients achieving a CR/CRu (complete response unconfirmed) as best response during induction treatment, rituximab maintenance treatment significantly prolonged the median disease free survival (DFS) compared to the observation group (53.7 versus 16.5 months, p = 0.0003) log-rank test (Table 4). The risk of relapse in complete responders was reduced by 67% (95% CI; 39% - 82%). (See Table 4.)

Click on icon to see table/diagram/image

The benefit of rituximab maintenance treatment was confirmed in all subgroups analysed, regardless of induction regimen (CHOP or R-CHOP) or quality of response to induction treatment (CR or PR) (Table 2). Rituximab maintenance treatment significantly prolonged median PFS in patients responding to CHOP induction therapy (median PFS 37.5 months versus 11.6 months, p < 0.0001) as well as in those responding to R-CHOP induction (median PFS 51.9 months versus 22.1 months, p = 0.0071). Rituximab maintenance treatment also provided a clinically meaningful benefit in terms of overall survival for both patients responding to CHOP and patients responding to R-CHOP in the induction phase of the study.
Rituximab maintenance treatment provided consistent benefit in all subgroups tested gender, age (≤60 years, >60 years), stage (III, IV), WHO performance status (0 versus > 0), B symptoms (absent, present), bone marrow involvement (no versus yes), IPI (0 - 2 versus 3 - 5), FLIPI score (0 - 1, versus 2 versus 3 - 5), number of extra-nodal sites (0 - 1 versus > 1), number of nodal sites (< 5 versus ≥ 5), number of previous regimens (1 versus 2), best response to prior therapy (CR/PR versus NC/PD), haemoglobin (< 12 g/dL versus ≥12 g/dL), β2 microglobulin (< 3 mg/L versus ≥ 3 mg/L), LDH (elevated, not elevated) except for the small subgroup of patients with bulky disease.
Adult Diffuse large B-cell non-Hodgkin's lymphoma: In a randomized, open-label trial, a total of 399 previously untreated elderly patients (age 60 to 80 years) with diffuse large B-cell lymphoma received standard CHOP chemotherapy (cyclophosphamide 750 mg/m2, doxorubicin 50 mg/m2, vincristine 1.4 mg/m2 up to a maximum of 2 mg on day 1, and prednisolone 40 mg/m2/day on days 1 - 5) every 3 weeks for eight cycles, or rituximab 375 mg/m2 plus CHOP (R-CHOP). Rituximab was administered on the first day of the treatment cycle.
The final efficacy analysis included all randomized patients (197 CHOP, 202 R-CHOP), and had a median follow-up duration of approximately 31 months. The two treatment groups were well balanced in baseline characteristics and disease status. The final analysis confirmed that R-CHOP significantly increased the duration of event-free survival (the primary efficacy parameter, where events were death, relapse or progression of lymphoma, or institution of a new anti-lymphoma treatment) (p = 0.0001). Kaplan Meier estimates of the median duration of event-free survival were 35 months in the R-CHOP arm compared to 13 months in the CHOP arm, representing a risk reduction of 41%. At 24 months, estimates for overall survival were 68.2% in the R-CHOP arm compared to 57.4% in the CHOP arm. A subsequent analysis of the duration of overall survival, carried out with a median follow-up duration of 60 months, confirmed the benefit of R-CHOP over CHOP treatment (p = 0.0071), representing a risk reduction of 32%.
The analysis of all secondary parameters (response rates, progression-free survival, disease-free survival, duration of response) verified the treatment effect of R-CHOP compared to CHOP. The complete response rate after cycle 8 was 76.2% in the R-CHOP group and 62.4% in the CHOP group (p = 0.0028). The risk of disease progression was reduced by 46% and the risk of relapse by 51%.
In all patient subgroups (gender, age, age-adjusted IPI, Ann Arbor stage, ECOG, Beta 2 Microglobulin, LDH, Albumin, B-symptoms, Bulky disease, extranodal sites, bone marrow involvement), the risk ratios for event-free survival and overall survival (R-CHOP compared with CHOP) were less than 0.83 and 0.95; respectively. R-CHOP was associated with improvements in outcome for both high- and low-risk patients according to age-adjusted IPI.
Previously untreated and relapsed/refractory chronic lymphocytic leukaemia: In two open-label randomized trials, a total of 817 previously untreated patients and 552 patients with relapsed/refractory CLL were randomized to receive either FC chemotherapy (fludarabine 25 mg/m2, cyclophosphamide 250 mg/m2, days 1-3) every 4 weeks for 6 cycles or rituximab in combination with FC (R-FC). Rituximab was administered at a dosage of 375 mg/m2 during the first cycle one day prior to chemotherapy and at a dosage of 500 mg/m2 on day 1 of each subsequent treatment cycle. A total of 810 patients (403 R-FC, 407 FC) the first line study (Table 5 and Table 6 as follows) and 552 patients (276 R-FC, 276 FC) for the relapsed/refractory study (Table 7) were analyzed for efficacy.
In the first line study, after a median observation time of 20.7 months, the median progression-free survival (primary endpoint) was 40 months in the R-FC group and 32 months in the FC group (p < 0.0001, log-rank test) (Table 5). The analysis of overall survival showed an improved survival in favour of the R-FC arm (p = 0.0427, log-rank test). These results were confirmed with longer follow-up: after a median observation time of 48.1 months, the median PFS was 55 months in the R-FC group and 33 months in the FC group (p<0.0001, log-rank test) and overall survival analyses continued to show a significant benefit of R-FC treatment over FC chemotherapy alone (p=0.0319, log-rank test). The benefit in terms of PFS was consistently observed in most patient subgroups analyzed according to disease risk at baseline. (i.e. Binet stages A-C) and was confirmed with longer follow-up (see Table 6). (See Tables 5 and 6.)

Click on icon to see table/diagram/image


Click on icon to see table/diagram/image

In the relapsed/refractory study, the median progression-free survival (primary endpoint) was 30.6 months in the R-FC group and 20.6 months in the FC group (p = 0.0002, log-rank test). The benefit in terms of PFS was observed in almost all patient subgroups analyzed according to disease risk at baseline. A slight but not significant improvement in overall survival was reported in the R-FC compared to the FC arm. (See Table 7.)

Click on icon to see table/diagram/image

Results from other supportive studies using rituximab in combination with other chemotherapy regimens (including CHOP, FCM, PC, PCM, bendamustine and cladribine) for the treatment of CLL patients have also demonstrated high overall response rates with promising PFS rates without adding relevant toxicity to the treatment.
Pediatric population: A multicenter, open-label, randomized study of Lymphome Malin B (LMB) chemotherapy (corticosteroids, vincristine, cyclophosphamide, high-dose methotrexate, cytarabine, doxorubicin, etoposide and triple drug [methotrexate/cytarabine/corticosteroid] intrathecal therapy) alone or in combination with rituximab was conducted in pediatric patients with previously untreated advanced stage CD20 positive DLBCL/BL/BAL/BLL. Advanced stage is defined as Stage III with elevated LDH level ("B-high"), [LDH > twice the institutional upper limit of the adult normal values (> Nx2)] or any stage IV or BAL. Patients were randomized to receive either LMB chemotherapy or six intravenous infusions of rituximab at a dose of 375 mg/m2 BSA in combination with LMB chemotherapy (two during each of the two induction courses and one during each of the two consolidation courses) as per the LMB scheme. A total of 328 randomized patients were included in the efficacy analyses, of which one patient under 3 years of age received rituximab in combination with LMB chemotherapy.
The two treatment arms, LMB (LMB chemotherapy) and R-LMB (LMB chemotherapy with rituximab), were well balanced with regards to baseline characteristics. Patients had a median age of 7 and 8 years in the LMB arm and R-LMB arm, respectively. Approximately half of patients were in Group B (50.6% in the LMB arm and 49.4% in the R-LMB arm), 39.6% in Group C1 in both arms, and 9.8% and 11.0% were in Group C3 in the LMB and R-LMB arms, respectively. Based on Murphy staging, most patients were either BL stage III (45.7% in the LMB arm and 43.3% in the R-LMB arm) or BAL, CNS negative (21.3% in the LMB arm and 24.4% in the R-LMB arm). Less than half of the patients (45.1% in both arms) had bone marrow involvement, and most patients (72.6% in the LMB arm and 73.2% in the R-LMB arm) had no CNS involvement. The primary efficacy endpoint was EFS, where an event was defined as occurrence of progressive disease, relapse, second malignancy, death from any cause, or non-response as evidenced by detection of viable cells in residue after the second CYVE course, whichever occurs first. The secondary efficacy endpoints were OS and CR (complete remission).
At the pre-specified interim analysis with approximately 1 year of median follow-up, clinically relevant improvement in the primary endpoint of EFS was observed, with 1-year rate estimates of 94.2% (95% CI, 88.5% - 97.2%) in the R-LMB arm vs. 81.5% (95% CI, 73.0% - 87.8%) in the LMB arm, and adjusted Cox HR 0.33 (95% CI, 0.14 - 0.79). Upon IDMC (independent data monitoring committee) recommendation based on this result, the randomization was halted and patients in the LMB arm were allowed to cross over to receive rituximab.
Primary efficacy analyses were performed in 328 randomized patients with median follow-up of 3.1 years. The results are described in Table 8. (See Table 8.)

Click on icon to see table/diagram/image

The primary efficacy analysis showed an EFS benefit of rituximab addition to LMB chemotherapy over LMB chemotherapy alone, with an EFS HR 0.32 (90% CI 0.17 - 0.58) from a Cox regression analysis adjusting for national group, histology, and therapeutic group. While no major differences in numbers of patients achieving CR was observed between the two treatment groups, the benefit of rituximab addition to LMB chemotherapy was also shown in the secondary endpoint of OS, with the OS HR of 0.36 (95% CI, 0.16 - 0.81).
Rheumatoid arthritis: The efficacy of rituximab in rheumatoid arthritis has been demonstrated in three pivotal, phase III, randomized, placebo-controlled, double-blind, multicenter studies. Eligible patients had severe active RA, diagnosed according to the criteria of the American College of Rheumatology (ACR). Rituximab was administered as two IV infusions separated by an interval of 15 days. Each course was preceded by an IV infusion of 100 mg methylprednisolone. All patients received concomitant oral methotrexate. In addition, in Study WA17042, all patients received concomitant oral glucocorticoid on days 2 - 7 and on days 8 to 14 following the first infusion.
The retreatment criteria differed between the studies using one of two approaches; 'Treatment to Remission' whereby patients were treated no more frequently than every 6 months if not in DAS28 remission (i.e. DAS28-ESR≥2.6) and 'Treatment as Needed' strategy ('Treatment PRN'), based on disease activity and/or return of clinical symptoms (swollen and tender joint counts ≥ 8) and treated no sooner than every 16 weeks.
Study WA17042 (REFLEX) included 517 patients that had experienced an inadequate response or intolerance to one or more TNF inhibitor therapies (TNF-IR). The primary endpoint was the proportion of patients who achieved an ACR20 response at week 24. Patients received 2 x 1000 mg rituximab or placebo. Patients were followed beyond week 24 for long term endpoints, including radiographic assessment at 56 weeks. During this time patients could receive further courses of rituximab under an open label extension study protocol. In the open-label protocol patients received further courses based on the 'Treatment PRN' criteria. Study WA17045 (SERENE) included 511 patients that had experienced an inadequate response to methotrexate (MTX-IR) and had not received prior biologic therapy. The primary endpoint was the proportion of patients who achieved an ACR20 response at week 24. Patients received either placebo, 2 x 500 mg or 2 x 1000 mg rituximab infusion. Patients were followed beyond week 24 for long term endpoints and could receive further courses of rituximab based on the 'Treatment to Remission' criteria. An active dose comparison was made at week 48.
Disease Activity Outcomes: In these studies, rituximab (2 x 1000 mg) significantly increased the proportion of patients achieving at least a 20% improvement in ACR score compared with patients treated with methotrexate alone (Table 9). Across all development studies the treatment benefit was similar in patients independent of age, gender, body surface area, race, number of prior treatments or disease status. Patients seropositive for disease related auto-antibodies (RF and/or anti CCP) demonstrated consistently high efficacy compared to MTX alone across studies. Efficacy in seropositive patients was higher than that observed in seronegative patients in whom efficacy was modest.
Clinically and statistically significant improvement was also noted on all individual components of the ACR response (tender and swollen joint counts, patient and physician global assessment, disability index scores (HAQ), pain assessment and CRP (mg/dL)). (See Table 9.)

Click on icon to see table/diagram/image

Patients treated with rituximab had a significantly greater reduction in disease activity score (DAS28) than patients treated with methotrexate alone. A good to moderate EULAR response was achieved by significantly more rituximab treated patients compared to patients treated with methotrexate alone (Table 10). (See Table 10.)

Click on icon to see table/diagram/image

Inhibition of Joint Damage: In studies WA17042 and WA17047 structural joint damage was assessed radiographically and expressed as change in modified Total Sharp Score (TSS) and its components, the erosion score and joint space narrowing score.
In Study WA17042, conducted in TNF-IR patients receiving rituximab in combination with methotrexate demonstrated significantly less radiographic progression at 56 weeks than patients from the methotrexate alone group. A higher proportion of patients receiving rituximab also had no erosive progression over 56 weeks.
Study WA17047, conducted in MTX-naïve patients (755 patients with early rheumatoid arthritis of between 8 weeks to four years duration), assessed the prevention of structural joint damage as its primary objective [see General: Rheumatoid Arthritis Patients (RA) Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA) Patients and Pemphigus Vulgaris (PV) Patients under Precautions]. Patients received either placebo, 2 x 500 mg or 2 x 1000 mg rituximab infusion. From week 24 patients could receive further courses of rituximab (or placebo to Week 104) based on the 'Treatment to Remission' criteria. The primary endpoint of change in modified Total Sharp Score (TSS) demonstrated that only treatment with rituximab at a dose of 2 x 1000 mg in combination with methotrexate significantly reduced the rate of progression of joint damage (PJD) at 52 weeks compared with placebo + MTX (Table 11). The reduction in PJD was driven mainly by a significant reduction in the change in Erosion Score.
Inhibition of the rate of progressive joint damage was also observed long term. Radiographic analysis at 2 years in study WA17042 demonstrated significantly reduced progression of structural joint damage in patients receiving rituximab (2 x 1000 mg) + MTX compared to MTX alone as well as a significantly higher proportion of patients with no progression of joint damage over the 2 year period. (See Table 11.)

Click on icon to see table/diagram/image

Quality of Life Outcomes: Rituximab treated patients reported an improvement in all patient-reported outcomes (HAQ-DI, FACIT-Fatigue and SF-36 questionnaires). Significant reductions in disability index (HAQ-DI), fatigue (FACIT-Fatigue), and improvement in the physical health domain of the SF-36 were observed in patients treated with rituximab compared to patients treated with methotrexate alone. (See Tables 12 and 13.)

Click on icon to see table/diagram/image


Click on icon to see table/diagram/image

Laboratory Evaluations: In rheumatoid factor (RF) positive patients, marked decreases were observed in rheumatoid factor concentrations following treatment with rituximab in all three studies (range 45 - 64%, Figure 1). (See Figure 1.)

Click on icon to see table/diagram/image

Plasma total immunoglobulin concentrations, total lymphocytes counts, and white cells generally remained within normal limits following rituximab treatment, with the exception of a transient drop in white cells counts over the first four weeks following therapy. Titers of IgG antigen specific antibody to mumps, rubella, varicella, tetanus toxoid, influenza and streptococcus pneumococci remained stable over 24 weeks following exposure to rituximab in rheumatoid arthritis patients.
Effects of rituximab on a variety of biomarkers was evaluated in patients enrolled into a clinical study. This substudy evaluated the impact of a single treatment course of rituximab on levels of biochemical markers, including markers of inflammation (Interleukin 6, C Reactive protein, Serum amyloid type A protein, Protein S100 isotypes A8 and A9), autoantibody (RF and anti-cyclic citrullinated peptide immunoglobulin) production and bone turnover (osteocalcin and procollagen 1 N terminal peptide (P1NP)). Rituximab treatment, whether as monotherapy or in combination with methotrexate or cyclophosphamide reduced the levels of inflammatory markers significantly, relative to methotrexate alone, over the first 24 weeks of follow-up. Levels of markers of bone turnover, osteocalcin and P1NP, increased significantly in the rituximab groups compared to methotrexate alone.
Long-term Efficacy with Multiple Course Therapy: In clinical studies patients were retreated based on either a 'Treatment to Remission' or a 'Treatment PRN' strategy. Repeat courses of rituximab maintained or improved treatment benefit, irrespective of the treatment strategy (Treatment to Remission or Treatment PRN) (Figure 2). However, Treatment to Remission generally provided better responses and tighter control of disease activity as indicated by ACRn, DAS28-ESR and HAQ-DI scores over time. Patients treated PRN also experienced returning disease symptoms between courses, as evidenced by DAS28-ESR scores which were close to pre-treatment levels prior to each course (Table 14). (See Table 14 and Figure 2.)

Click on icon to see table/diagram/image


Click on icon to see table/diagram/image

Adult Patients with Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA): Adult Induction of Remission (GPA/MPA Study 1): A total of 197 patients with severe, active Granulomatosis with polyangiitis (Wegener's) (GPA) and Microscopic polyangiitis (MPA) were enrolled and treated in an active controlled, randomized, double-blind, multicenter, non-inferiority study. Patients were 15 years of age or older, diagnosed with severely, active Granulomatosis with polyangiitis (Wegener's) (75% of patients) or Microscopic Polyangiitis (MPA) (24% of patients) according to the Chapel Hill Consensus conference criteria (1% of patients had unknown GPA and MPA type). Patients were randomized in a 1:1 ratio to receive either oral cyclophosphamide daily (2 mg/kg/day) for 3 to 6 months, followed by azathioprine or rituximab (375 mg/m2) once weekly for 4 weeks. Patients in both arms received 1000 mg of pulse intravenous methylprednisolone (or another equivalent-dose glucocorticoid) per day for 1 to 3 days, followed by oral prednisone (1 mg/kg/day, not exceeding 80 mg/day). Prednisone tapering was to be completed by 6 months from the start of study treatment.
The primary outcome measure was achievement of complete remission at 6 months defined as a Birmingham Vasculitis Activity Score for Wegener's Granulomatosis (BVAS/WG) of 0, and off glucocorticoid therapy. The prespecified non-inferiority margin for the treatment difference was 20%. The study demonstrated non-inferiority of rituximab to cyclophosphamide for complete remission at 6 months (Table 15). Efficacy was observed both for patients with newly diagnosed GPA and MPA and for patients with relapsing disease. (Table 16). (See Tables 15 and 16.)

Click on icon to see table/diagram/image


Click on icon to see table/diagram/image

Complete remission at 12 and 18 months: In the rituximab group, 48% of patients achieved CR at 12 months, and 39% of patients achieved CR at 18 months. In patients treated with cyclophosphamide (followed by azathioprine for maintenance of complete remission), 39% of patients achieved CR at 12 months, and 33% of patients achieved CR at 18 months. From month 12 to month 18, 8 relapses were observed in the rituximab group compared with four in the cyclophosphamide group.
Laboratory evaluations: A total of 23/99 (23%) rituximab-treated patients from the induction of remission trial tested positive for ADA by 18 months. None of the 99 rituximab-treated patients were ADA positive at screening. There was no apparent trend or negative impact of the presence of ADA on safety or efficacy in the induction of the remission trial.
Adult Maintenance Treatment (GPA/MPA Study 2): A total of 117 patients (88 with GPA, 24 with MPA, and 5 with renal-limited ANCA-associated vasculitis) in disease remission were randomized to receive azathioprine (59 patients) or rituximab (58 patients) in this prospective, multi-center, controlled, open-label study. Eligible patients were 21 to 75 years of age and had newly diagnosed or relapsing disease in complete remission after combined treatment with glucocorticoids and pulse cyclophosphamide. Patients were ANCA-positive at diagnosis or during the course of their disease; had histologically confirmed necrotizing small-vessel vasculitis with a clinical phenotype of GPA/MPA, or renal limited ANCA-associated vasculitis; or both. Remission-induction therapy included IV prednisone, administered as per the investigator's discretion, preceded in some patients by methylprednisolone pulses, and pulse cyclophosphamide until remission was attained after 4 to 6 months. At that time, and within a maximum of 1 month after the last cyclophosphamide pulse, patients were randomly assigned to receive either rituximab (two 500 mg IV infusion separated by two weeks (on Day 1 and Day 15) followed by 500 mg IV every 6 months for 18 months or azathioprine (administered orally at a dose of 2 mg/kg/day for 12 months, then 1.5 mg/kg/day for 6 months, and finally 1 mg/kg/day for 4 months (treatment discontinuation after these 22 months). Prednisone treatment was tapered and then kept at a low dose (approximately 5 mg per day) for at least 18 months after randomization. Prednisone dose tapering and the decision to stop prednisone treatment after month 18 were left at the investigator's discretion.
All patients were followed until month 28 (10 or 6 months, respectively, after the last rituximab infusion or azathioprine dose). Pneumocystis jirovecii pneumonia prophylaxis was required for all patients with CD4+ T-lymphocyte counts less than 250 per cubic millimeter.
The primary outcome measure was the rate of major relapse at month 28.
Results: At month 28, major relapse (defined by the reappearance of clinical and/or laboratory signs of vasculitis activity ([BVAS]>0) that could lead to organ failure or damage or could be life threatening) occurred in three patients (5%) in the rituximab group and 17 patients (29%) in the azathioprine group (p=0.0007). Adjusting for the stratification factor using Cox PH modeling, rituximab reduced the risk of major relapse by approximately 86% relative to azathioprine (hazard ratio [HR]: 0.14; 95% confidence interval [CI]: 0.04, 0.47). Minor relapses (not life threatening and not involving major organ damage) occurred in seven patients in the rituximab group (12%) and eight patients in the azathioprine group (14%).
The cumulative incidence rate curves showed that time to first major relapse was longer in patients with rituximab starting from Month 2 and was maintained up to Month 28 (Figure 3). (See Figure 3.)

Click on icon to see table/diagram/image

Laboratory evaluations: A total of 6/34 (18%) of rituximab treated patients from the maintenance therapy clinical trial developed ADA. There was no apparent trend or negative impact of the presence of ADA on safety or efficacy in the maintenance therapy clinical trial.
Pediatric Patients with Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA): Study WA25615 (PePRS) was a multicenter, open-label, single-arm, uncontrolled study in 25 patients (≥2 to <18 years old) with active GPA/MPA. The median age of patients in the study was: 14 years (range: 6-17 years) and the majority of patients (20/25 [80%]) were female. A total of 19 patients (76%) had GPA and 6 patients (24%) had MPA at baseline. Eighteen patients (72%) had newly diagnosed disease upon study entry (13 patients with GPA and 5 patients with MPA) and 7 patients had relapsing disease (6 patients with GPA and 1 patient with MPA).
The study design consisted of an initial 6-month remission induction phase, and a minimum 18-month follow-up phase up to a maximum of 54 months (4.5 years). The remission induction regimen consisted of four once weekly IV infusions of rituximab at a dose of 375 mg/m2 BSA, on study days 1, 8, 15 and 22 in combination with oral prednisolone or prednisone at 1 mg/kg/day (max 60 mg/day) tapered to 0.2 mg/kg/day minimum (max 10 mg/day) by month 6. After the remission induction phase, patients could receive subsequent rituximab infusion on or after month 6 to maintain remission and control disease activity. Patients were to receive a minimum of 3 doses of IV methylprednisolone (30 mg/kg/day, not exceeding 1 g/day) prior to the first rituximab infusion. If clinically indicated, additional daily doses (up to three) of IV methylprednisolone could be given.
All 25 patients completed all four once weekly IV infusions for the 6-month remission induction phase. A total of 24 out of 25 patients completed at least 18 months of follow-up.
The objectives of this study was to evaluate safety, PK parameters, and efficacy of rituximab in pediatric GPA/MPA patients (≥2 to <18 years old). The efficacy objectives of the study were exploratory and principally assessed using the Pediatric Vasculitis Activity Score (PVAS) (Table 17). (See Table 17.)

Click on icon to see table/diagram/image

Cumulative Glucocorticoid dose (IV and Oral) by Month 6: A clinically meaningful decrease in median overall oral glucocorticoid was observed from week 1 (median=45 mg prednisone equivalent dose [IQR: 35-60]) to month 6 (median=7.5 mg [IQR: 4-10]), which was subsequently maintained at month 12 (median - 5 mg [IQR: 2-10]) and month 18 (median=5 mg [IQR: 1-5]).
Follow-Up Treatment: After the 6-month remission induction phase, patients who had not achieved remission or who had progressive disease or flare that could not be controlled by glucocorticoids alone received additional treatment for GPA/MPA, that could include rituximab and/or other therapies, at the discretion of the investigator.
Fourteen out of 25 patients (56%) received additional rituximab treatment at or post month 6, up to month 18. Five patients received four once weekly doses (375 mg/m2) of rituximab approximately every 6 months; 5 patients received a single dose (375 mg/m2) of rituximab every 6 months, and a further 4 patients received various other rituximab doses/regimens according to their treating physician. Of the 14 patients, 9 patients achieved PVAS remission by month 6 and sustained remission through month 18: 4 patients achieved remission between month 6 and 12 and sustained remission through month 18. One patient first achieved remission between month 12 and 18.
Laboratory evaluations: A total of 4/25 patients (16%) developed ADA during the overall study period. Limited data shows there was no trend observed in the adverse reactions reported in ADA positive patients.
There was no apparent trend or negative impact of the presence of ADA on safety or efficacy in the pediatric GPA and MPA clinical trials.
Pemphigus Vulgaris: PV Study 1 (Study ML22196): The efficacy and safety of rituximab in combination with short-term low dose glucocorticoid (prednisone) therapy were evaluated in newly diagnosed patients with moderate to severe pemphigus (74 pemphigus vulgaris [PV] and 16 pemphigus foliaceus [PF]) in this randomized, open-label, controlled, multicenter study. Patients were between 19 and 79 years of age and had not received prior therapies for pemphigus. In the PV population, five (13%) patients in the rituximab group and three (8%) patients in the standard prednisone group had moderate disease and 33 (87%) patients in the rituximab group and 33 (92%) in the standard dose prednisone group had severe disease according to disease severity defined by Harman's criteria.
Patients were stratified by baseline disease severity (moderate or severe) and randomized 1:1 to receive either rituximab and low dose prednisone or standard dose prednisone. Patients randomized to the rituximab group received an initial intravenous infusion of 1000 mg rituximab on Study Day 1 in combination with 0.5 mg/kg/day oral prednisone tapered off over 3 months if they had moderate disease or 1 mg/kg/day oral prednisone tapered off over 6 months if they had severe disease, and a second intravenous infusion of 1000 mg on Study Day 15. Maintenance infusions of rituximab 500 mg were administered at months 12 and 18. Patients randomized to the standard dose prednisone group received an initial 1 mg/kg/day oral prednisone tapered off over 12 months if they had moderate disease or 1.5 mg/kg/day oral prednisone tapered off over 18 months if they had severe disease. Patients in the rituximab group who relapsed could receive an additional infusion of rituximab 1000 mg in combination with reintroduced or escalated prednisone dose. Maintenance and relapse infusions were administered no sooner than 16 weeks following the previous infusion.
The primary objective for the study was complete remission (complete epithelialization and absence of new and/or established lesions) at month 24 without the use of prednisone therapy for two months or more (CRoff for ≥2 months). Other efficacy parameters included evaluation of severe and moderate relapses (severity as defined by Harman's criteria and relapse defined as the appearance of ≥3 new lesions a month that did not heal spontaneously within 1 week, or the extension of established lesions in a patient who had achieved disease control), evaluation of the total median cumulative dose of prednisone, and the median duration of complete remission off corticosteroid therapy.
PV study 1 Results: The study demonstrated superiority of rituximab and low dose prednisone over standard dose prednisone in achieving CRoff ≥ 2 months at month 24 in PV patients (see Table 18). Additionally, at month 24, the proportion of PV patients with CRoff ≥ 3 months was higher in the rituximab and low dose prednisone group compared to the standard dose prednisone group (34 patients [90%] vs. 9 patients [25%], p value <0.0001). (See Table 18.)

Click on icon to see table/diagram/image

Rituximab was considered steroid-sparing based on the duration that PV patients were off glucocorticoid therapy and cumulative exposure to glucocorticoids in the rituximab group compared to the standard dose prednisone group.
Duration off Glucocorticoid Therapy: Of PV patients who responded at month 24, the median duration of CRoff ≥ 2 months in the rituximab group was 498.5 days compared to 125 days in the standard dose prednisone group.
Glucocorticoid Exposure: The median (min, max) cumulative prednisone dose at month 24 was 5800 mg (2304, 29303) in the rituximab group compared to 20520 mg (2409, 60565) in the standard dose prednisone group.
Severe or Moderate Relapses: At month 24, 9 (24%) PV patients in the rituximab group experienced at least one severe or moderate relapse vs. 18 (50%) PV patients in the standard dose prednisone group.
PV Study 2 (Study WA29330): In a randomized, double-blind, double-dummy, active-comparator, multicenter study, the efficacy and safety of rituximab compared with mycophenolate mofetil (MMF) were evaluated in patients with moderate-to-severe PV receiving 60-120 mg/day oral prednisone or equivalent (1.0-1.5 mg/kg/day) at study entry and tapered to reach a dose of 60 or 80 mg/day by Day 1. Patients had a confirmed diagnosis of PV within the previous 24 months and evidence of moderate-to-severe disease (defined as a total Pemphigus Disease Area Index, PDAI, activity score of ≥ 15).
One hundred and thirty-five patients were randomized to treatment with rituximab 1000 mg administered on Day 1, Day 15, Week 24 and Week 26 or oral MMF 2 g/day for 52 weeks in combination with 60 or 80 mg oral prednisone with the aim of tapering to 0 mg/day prednisone by Week 24.
The primary efficacy objective for this study was to evaluate at week 52, the efficacy of rituximab compared with MMF in achieving sustained complete remission defined as achieving healing of lesions with no new active lesions (i.e., PDAI activity score of 0) while on 0 mg/day prednisone or equivalent, and maintaining this response for at least 16 consecutive weeks, during the 52-week treatment period.
PV Study 2 Results: The study demonstrated the superiority of rituximab over MMF in combination with a tapering course of oral corticosteroids in achieving CR off corticosteroid ≥ 16 weeks at Week 52 in PV patients (Table 19). The majority of patients in the mITT population were newly diagnosed (74%) and 26% of patients had established disease (duration of illness ≥ 6 months and received prior treatment for PV). (See Table 19.)

Click on icon to see table/diagram/image

The analysis of all secondary parameters (including cumulative oral corticosteroid dose, the total number of disease flares, and change in health-related quality of life, as measured by the Dermatology Life Quality Index) verified the statistically significant results of rituximab compared to MMF. Testing of secondary endpoints were controlled for multiplicity.
Glucocorticoid exposure: The cumulative oral corticosteroid dose was significantly lower in patients treated with rituximab. The median (min, max) cumulative prednisone dose at Week 52 was 2775 mg (450, 22180) in the rituximab group compared to 4005 mg (900, 19920) in the MMF group (p=0.0005).
Disease flare: The total number of disease flares was significantly lower in patients treated with rituximab compared to MMF (6 vs. 44, p<0.0001) and there were fewer patients who had at least one disease flare (8.1% vs. 41.3%).
Laboratory evaluations: By week 52, a total of 20/63 (31.7%) (19 treatment-induced and 1 treatment-enhanced) rituximab-treated PV patients tested positive for ADA. There was no apparent negative impact of the presence of ADA on safety or efficacy in PV Study 2.
IMMUNOGENICITY: As with all therapeutic patients, there is the potential for an immune response in patients treated with rituximab. The data reflects the number of patients whose test results were considered positive for antibodies to rituximab using an enzyme-linked immunosorbent assay (ELISA). Immunogenicity assay results may be influenced by several factors including assay sensitivity and specificity, sample handling, timing of sample collection, concomitant medicinal products and underlying disease. For these reasons, comparison of incidence of antibodies to rituximab with the incidence of antibodies in other studies or to other products may be misleading.
Rheumatoid Arthritis: Approximately 10% of patients with rheumatoid arthritis tested positive for anti-drug antibodies (ADA) in the RA clinical studies. The emergence of ADA was not associated with clinical deterioration or with an increased risk of reactions to subsequent infusions in the majority of patients. The presence of ADA may be associated with worsening of infusion or allergic reactions after the second infusion of subsequent courses, and failure to deplete B cells after receipt of further treatment courses has been observed rarely.
Adult and Pediatric Patients with Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA): Twenty-three percent (23/99) of rituximab treated patients from the adult GPA and MPA induction of remission trial and 18% (6/34) of rituximab treated patients in the maintenance therapy clinical trial developed ADA.
In the pediatric clinical trial, a total of 4/25 patients (16%) developed ADA during the overall study period. Limited data shows there was no trend observed in the adverse reactions reported in ADA positive patients. There was no apparent trend or negative impact of the presence of ADA on safety or efficacy in the adult and pediatric GPA and MPA clinical trials.
Pemphigus Vulgaris: By 18 months, a total of 19/34 (56%) (14 treatment-induced and 5 treatment-enhanced) rituximab treated PV patients tested positive for ADA. There was no apparent negative impact of the presence of ADA on safety or efficacy in the PV clinical study.
Pharmacokinetics: Distribution: Non-Hodgkin's Lymphoma: Based on a population pharmacokinetic analysis in 298 NHL patients who received single or multiple infusions of rituximab as a single agent or in combination with CHOP therapy, the typical population estimates of nonspecific clearance (CL1), specific clearance (CL2) likely contributed by B cells or tumour burden, and central compartment volume of distribution (V1) were 0.14 L/day, 0.59 L/day, and 2.7 L, respectively. The estimated median terminal elimination half-life of rituximab was 22 days (range, 6.1 to 52 days). Baseline CD19-positive cell counts and size of measurable tumour lesions contributed to some of the variability in CL2 of rituximab in data from 161 patients given 375 mg/m2 as an IV infusion for 4 weekly doses. Patients with higher CD19-positive cell counts or tumour lesions had a higher CL2. However, a large component of inter-individual variability remained for CL2 after correction for CD19-positive cell counts and tumour lesion size. V1 varied by body surface area (BSA) and CHOP therapy. This variability in V1 (27.1% and 19.0%) contributed by the range in BSA (1.53 to 2.32 m2) and concurrent CHOP therapy, respectively, were relatively small. Age, gender, race, and WHO performance status had no effect on the pharmacokinetics of rituximab. This analysis suggests that dose adjustment of rituximab with any of the tested covariates is not expected to result in a meaningful reduction in its pharmacokinetic variability.
Rituximab at a dose of 375 mg/m2 was administered as an IV infusion at weekly intervals for 4 doses to 203 patients with NHL naïve to rituximab. The mean Cmax following the fourth infusion was 486 µg/mL (range, 77.5 to 996.6 µg/mL). The peak and trough serum levels of rituximab were inversely correlated with baseline values for the number of circulating CD19-positive B-cells and measures of disease burden. Median steady-state serum levels were higher for responders compared with non-responders. Serum levels were higher in patients with International Working Formulation (IWF) subtypes B, C, and D as compared with those with subtype A. Rituximab was detectable in the serum of patients 3 - 6 months after completion of last treatment.
Rituximab at a dose of 375 mg/m2 was administered as an IV infusion at weekly intervals for 8 doses to 37 patients with NHL. The mean Cmax increased with each successive infusion, spanning from a mean of 243 µg/mL (range, 16 - 582 µg/mL) after the first infusion to 550 µg/mL (range, 171 - 1177 µg/mL) after the eighth infusion.
The pharmacokinetic profile of rituximab when administered as 6 infusions of 375 mg/m2 in combination with 6 cycles of CHOP chemotherapy was similar to that seen with rituximab alone.
Pediatric DLBCL/BL/BAL/BLL: In the clinical trial studying paediatric DLBCL/BL/BAL/BLL, the PK was studied in a subset of 35 patients aged 3 years and older. The PK was comparable between the two age groups (≥3 to <12 years vs. ≥12 to <18 years). After two rituximab intravenous infusions of 375 mg/m2 in each of the two induction cycles (cycle 1 and 2) followed by one rituximab intravenous infusion of 375 mg/m2 in each of the consolidation cycles (cycle 3 and 4) the maximum concentration was highest after the fourth infusion (cycle 2) with a geometric mean of 347 µg/mL followed by lower geometric mean maximum concentrations thereafter (Cycle 4: 247 µg/mL). With this dose regimen, trough levels were sustained (geometric means: 41.8 µg/mL (pre-dose Cycle 2; after 1 cycle), 67.7 µg/mL (pre-dose Cycle 3, after 2 cycles) and 58.5 µg/mL (pre-dose Cycle 4, after 3 cycles)). The median elimination half-life in paediatric patients aged 3 years and older was 26 days.
The PK characteristics of rituximab in paediatric patients with DLBCL/BL/BAL/BLL were similar to what has been observed in adult NHL patients.
No PK data are available in the ≥ 6 months to < 3 years age group, however, population PK prediction supports comparable systemic exposure (AUC, Ctrough) in this age group compared to ≥ 3 years (Table 19). Smaller baseline tumor size is related to higher exposure due to lower time dependent clearance, however, systemic exposures impacted by different tumor sizes remain in the range of exposure that was efficacious and had an acceptable safety profile. (See Table 20.)

Click on icon to see table/diagram/image

Chronic Lymphocytic Leukaemia: Rituximab was administered as an IV infusion at a first-cycle dose of 375 mg/m2 increased to 500 mg/m2 each cycle for 5 doses in combination with fludarabine and cyclophosphamide in CLL patients. The mean Cmax (N = 15) was 408 µg/mL (range, 97 - 764 µg/mL) after the fifth 500 mg/m2 infusion.
Rheumatoid Arthritis: Following two intravenous infusions of rituximab at a dose of 1000 mg, two weeks apart, the mean terminal half-life was 20.8 days (range, 8.58 to 35.9 days), mean systemic clearance was 0.23 L/day (range, 0.091 to 0.67 L/day), and mean steady-state distribution volume was 4.6 L (range, 1.7 to 7.51 L). Population pharmacokinetic analysis of the same data gave similar mean values for systemic clearance and half-life, 0.26 L/day and 20.4 days, respectively. Population pharmacokinetic analysis revealed that BSA and gender were the most significant covariates to explain inter individual variability in pharmacokinetic parameters. After adjusting for BSA, male subjects had a larger volume of distribution and a faster clearance than female subjects. The gender-related pharmacokinetic differences are not considered to be clinically relevant and dose adjustment is not required.
The pharmacokinetics of rituximab were assessed following two IV doses of 500 mg and 1000 mg on Days 1 and 15 in four studies. In all these studies, rituximab pharmacokinetics were dose proportional over the limited dose range studied. Mean Cmax for serum rituximab following first infusion ranged from 157 to 171 µg/mL for 2 x 500 mg dose and ranged from 298 to 341 μg/mL for 2 x 1000 mg dose. Following second infusion, mean Cmax ranged from 183 to 198 μg/mL for the 2 x 500 mg dose and ranged from 355 to 404 μg/mL for the 2 x 1000 mg dose. Mean terminal elimination half-life ranged from 15 to 16.5 days for the 2 x 500 mg dose group and 17 to 21 days for the 2 x 1000 mg dose group. Mean Cmax was 16 to 19% higher following second infusion compared to the first infusion for both doses.
The pharmacokinetics of rituximab were assessed following two IV doses of 500 mg and 1000 mg upon re-treatment in the second course. Mean Cmax for serum rituximab following first infusion was 170 to 175 μg/mL for 2 x 500 mg dose and 317 to 370 μg/mL for 2 x 1000 mg dose. Cmax following second infusion, was 207 μg/mL for the 2 x 500 mg dose and ranged from 377 to 386 μg/mL for the 2 x 1000 mg dose. Mean terminal elimination half-life after the second infusion, following the second course, was 19 days for 2 x 500 mg dose and ranged from 21 to 22 days for the 2 x 1000 mg dose. PK parameters for rituximab were comparable over the two treatment courses.
The pharmacokinetic parameters in the anti-TNF inadequate responder population, following the same dosage regimen (2 x 1000 mg, iv, 2 weeks apart), were similar with a mean maximum serum concentration of 369 μg/mL and a mean terminal half-life of 19.2 days.
Adult and Pediatric Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA): The PK parameter in adult and pediatric patients with GPA/MPA receiving 375 mg/m2 rituximab once weekly for four doses are summarized in Table 21. (See Table 21.)

Click on icon to see table/diagram/image

The PK parameters of rituximab in adult GPA/MPA patients appear similar to what has been observed in RA patients (see Distribution as previously mentioned).
Based on a population pharmacokinetic analysis in pediatric patients with GPA/MPA, the PK parameters of rituximab were similar to those in adults with GPA/MPA, once taking into account the BSA effect on clearance and volume parameters.
Pemphigus vulgaris: The PK parameters in adult PV patients receiving rituximab 1000 mg at Days 1, 15, 168, and 182 are summarized in Table 22. (See Table 22.)

Click on icon to see table/diagram/image

Following the first two rituximab administrations (at day 1 and 15, corresponding to cycle 1), the PK parameters of rituximab in patients with PV were similar to those in patients with GPA/MPA and patients with RA.
Following the last two administrations (at day 168 and 182, corresponding to cycle 2), rituximab clearance decreased while the central volume of distribution remained unchanged.
Elimination: See Distribution as previously mentioned.
Pharmacokinetics in Special Populations: Renal impairment: No pharmacokinetic data are available in patients with renal impairment.
Hepatic impairment: No pharmacokinetic data are available in patients with hepatic impairment.
Pediatrics: The effect of body surface area on the pharmacokinetics of rituximab was assessed in a population pharmacokinetic analysis which included 9 children (≥ 6 years to <12 years) and 16 adolescents (12 to <18 years) with GPA/MPA. BSA was a significant covariate on rituximab pharmacokinetics (see Special Dosage Instructions under Dosage & Administration).
Indications/Uses
Non-Hodgkin's Lymphoma: Truxima is indicated for the treatment of: patients with relapsed or chemoresistant low-grade or follicular, CD20-positive, B-cell non-Hodgkin's lymphoma; previously untreated patients with stage III-IV follicular lymphoma in combination with chemotherapy; patients with follicular lymphoma as maintenance treatment, after response to induction therapy; patients with CD20-positive diffuse large B-cell non-Hodgkin's lymphoma in combination with CHOP (cyclophosphamide, doxorubicin, vincristine and prednisone) chemotherapy.
Truxima in combination with chemotherapy is indicated for the treatment of pediatric patients (aged ≥ 6 months to < 18 years old) with previously untreated advanced stage CD20 positive diffuse large B-cell lymphoma (DLBCL), Burkitt lymphoma (BL)/Burkitt leukaemia (mature B-cell acute leukaemia) (BAL) or Burkitt-like lymphoma (BLL).
Chronic Lymphocytic Leukaemia: Truxima in combination with chemotherapy is indicated for the treatment of patients with previously untreated and relapsed/refractory chronic lymphocytic leukaemia (CLL).
Rheumatoid Arthritis: Truxima in combination with methotrexate is indicated in adult patients for: the treatment of moderate to severe, active rheumatoid arthritis when the response to disease-modifying anti-rheumatic drugs including methotrexate has been inadequate; treatment of moderate to severe, active rheumatoid arthritis in patients with an inadequate response or intolerance to one or more tumour necrosis factor (TNF) inhibitor therapies.
Truxima has been shown to reduce the rate of progression of joint damage as measured by x-ray, to improve physical function and to induce major clinical response, when given in combination with methotrexate.
Adult and Pediatric Patients with Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA): Truxima in combination with glucocorticoids is indicated for the treatment of adult patients with severe active granulomatosis with polyangiitis (GPA, also known as Wegener's Granulomatosis) and microscopic polyangiitis (MPA) (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
Truxima, in combination with glucocorticoids, is indicated for the induction of remission in pediatric patients (aged ≥ 2 to < 18 years old) with severe, active GPA (Wegener's) and MPA.
Pemphigus Vulgaris: Truxima is indicated for the treatment of patients with moderate to severe pemphigus vulgaris (PV).
Dosage/Direction for Use
General: Substitution by any other biological medicinal product requires the consent of the prescribing physician.
It is important to check the product labels to ensure that the appropriate strength is being given to the patients, as prescribed.
Truxima should be administered as an IV infusion through a dedicated line, in an environment where full resuscitation facilities are immediately available and under the close supervision of an experienced physician.
Do not administer the prepared infusion solutions as an IV push or bolus.
Premedication and Prophylactic Medications: Premedication consisting of an analgesic/anti-pyretic (e.g. paracetamol/acetaminophen) and an anti-histaminic drug (e.g. diphenhydramine) should always be administered before each infusion of Truxima. Premedication with glucocorticoids should be administered in order to reduce the frequency and severity of infusion-related reactions.
In adult patients with non-Hodgkin's lymphoma and CLL, premedication with glucocorticoids should be considered if Truxima is not given in combination with glucocorticoid-containing chemotherapy. In pediatric patients with non-Hodgkin's lymphoma, premedication with paracetamol and H1 antihistamine (= diphenhydramine or equivalent) should be administered 30 to 60 minutes before the start of the infusion of Truxima. In addition, prednisone should be given as indicated in Table 23.
Prophylaxis with adequate hydration and administration of uricostatics starting 48 hours prior to start of therapy is recommended for CLL patients to reduce the risk of tumour lysis syndrome. For CLL patients whose lymphocyte counts are > 25 x 109/L it is recommended to administer prednisone/prednisolone 100 mg intravenously shortly before administration with Truxima to decrease the rate and severity of acute infusion reactions and/or cytokine release syndrome.
In patients with rheumatoid arthritis, GPA or MPA or pemphigus vulgaris, premedication with 100 mg intravenous methylprednisolone should be completed 30 minutes prior to each infusion of Truxima to decrease the incidence and severity of infusion related reactions (IRRs).
In adult patients with GPA or MPA, methylprednisolone given intravenously for 1 to 3 days at a dose of 1000 mg per day is recommended prior to the first infusion of Truxima (the last dose of methylprednisolone may be given on the same day as the first infusion of Truxima). This should be followed by oral prednisone 1 mg/kg/day (not to exceed 80 mg/day, and tapered as rapidly as possible based on clinical need) during and after the 4 week induction course of Truxima treatment.
Pneumocystis jirovecii pneumonia (PJP) prophylaxis is recommended for adult patients with GPA/MPA or PV during and following rituximab treatment, as appropriate according to local clinical practice guidelines.
Pediatric population: In pediatric patients with GPA or MPA, prior to the first Truxima intravenous infusion, methylprednisolone should be given intravenous for three daily doses of 30 mg/kg/day (not to exceed 1 g/day) to treat severe vasculitis symptoms. Up to three additional daily doses of 30 mg/kg intravenous methylprednisolone can be given prior to the first Truxima infusion.
Following completion of intravenous methylprednisolone administration, patients should receive oral prednisone 1 mg/kg/day (not to exceed 60 mg/day) and tapered as rapidly as possible per clinical need (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
Pneumocystis jirovecii pneumonia (PJP) prophylaxis is recommended for paediatric patients with GPA or MPA during and following rituximab treatment, as appropriate.
Dosage adjustments during treatment: No dose reductions of Truxima are recommended. When Truxima is given in combination with chemotherapy, standard dose reductions for the chemotherapeutic drugs should be applied.
Standard dosage: Low-grade or follicular non-Hodgkin's lymphoma: Initial treatment: The recommended dosage of Truxima used as monotherapy for adult patients is 375 mg/m2 body surface area (BSA), administered as an IV infusion (see "First infusion" and "Subsequent infusions" as follows) once weekly for 4 weeks.
The recommended dosage of Truxima in combination with any chemotherapy is 375 mg/m2 body surface area per cycle for a total of: 8 cycles with R-CVP (21 days/cycle); 8 cycles with R-MCP (28 days/cycle); 8 cycles with R-CHOP (21 days/cycle); 6 cycles if a complete remission is achieved after 4 cycles; 6 cycles with R-CHVP-Interferon (21 days/cycle).
Truxima should be administered on day 1 of each chemotherapy cycle after IV administration of the glucocorticoid component of the chemotherapy, if applicable.
Re-treatment following relapse: Patients who have responded to Truxima initially may receive Truxima at a dose of 375 mg/m2 body surface area, administered as an IV infusion once weekly for 4 weeks (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies: Low-grade or follicular non-Hodgkin's lymphoma: Monotherapy: Re-treatment, weekly for 4 doses under Actions).
Maintenance treatment: Previously untreated patients after response to induction treatment may receive maintenance therapy with Truxima given at 375 mg/m2 body surface area once every 2 months until disease progression or for a maximum period of two years (12 infusions).
Relapsed/refractory patients after response to induction treatment may receive maintenance therapy with Truxima given at 375 mg/m2 body surface area once every 3 months until disease progression or for a maximum period of two years (8 infusions in total).
Adult Diffuse large B-cell non-Hodgkin's lymphoma: In patients with diffuse large B cell non-Hodgkin's lymphoma. Truxima should be used in combination with CHOP (cyclophosphamide, doxorubicin, prednisone and vincristine) chemotherapy. The recommended dosage of Truxima is 375 mg/m2 body surface area, administered on day 1 of each chemotherapy cycle for 8 cycles after IV administration of the glucocorticoid component of CHOP. The other components of CHOP should be given after the administration of Truxima (see "First infusion" and "Subsequent infusions" as follows).
First infusion: The recommended initial infusion rate is 50 mg/h; subsequently, the rate can be escalated in 50 mg/h increments every 30 minutes to a maximum of 400 mg/h.
Subsequent infusions: Subsequent infusions of Truxima can be started at a rate of 100 mg/h and increased by 100 mg/h increments every 30 minutes to a maximum of 400 mg/h.
Pediatric non-Hodgkin's lymphoma: In pediatric patients from ≥ 6 months to < 18 years of age with previously untreated, advanced stage CD20 positive DLBCL/BL/BAL/BLL, Truxima should be used in combination with systemic Lymphome Malin B (LMB) chemotherapy (see Tables 23 and 24). The recommended dosage of Truxima is 375 mg/m2 BSA, administered as an intravenous infusion. No Truxima dose adjustments, other than by BSA, are required. (See Tables 23 and 24.)

Click on icon to see table/diagram/image


Click on icon to see table/diagram/image

First infusion: The recommended initial rate for infusion is 0.5 mg/kg/h (maximum 50 mg/h); it can be escalated by 0.5 mg/kg/h every 30 minutes if there is no hypersensitivity or infusion-related reactions, to a maximum of 400 mg/h.
Subsequent infusions: Subsequent doses of Truxima can be infused at an initial rate of 1 mg/kg/h (maximum 50 mg/h); it can be increased by 1 mg/kg/h every 30 minutes to a maximum of 400 mg/h.
Chronic Lymphocytic Leukaemia: The recommended dosage of Truxima in combination with chemotherapy for previously untreated and relapsed/refractory patients is 375 mg/m2 body surface area administered on day 1 of the first treatment cycle followed by 500 mg/m2 body surface area administered on day 1 of each subsequent cycle for 6 cycles in total (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions). The chemotherapy should be given after Truxima infusion.
First infusion: The recommended initial infusion rate is 50 mg/h; subsequently, the rate can be escalated in 50 mg/h increments every 30 minutes to a maximum of 400 mg/h.
Subsequent infusions: Subsequent infusions of Truxima can be started at a rate of 100 mg/h and increased by 100 mg/h increments every 30 minutes to a maximum of 400 mg/h.
Dosage adjustments during treatment: No dose reductions of Truxima are recommended. When Truxima is given in combination with chemotherapy, standard dose reductions for the chemotherapeutic drugs should be applied.
Rheumatoid arthritis (RA): A course of Truxima consists of two 1000 mg IV infusions. The recommended dosage of Truxima is 1000 mg by IV infusion followed two weeks later by the second 1000 mg IV infusion.
The need for further courses should be evaluated 24 weeks following the previous course with retreatment given based on residual or disease activity returning to a level above a DAS28-ESR of 2.6 (treatment to remission) (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions). Patients may receive further courses no sooner than 16 weeks following the previous course.
Patients should receive 100 mg IV methylprednisolone to be completed 30 minutes prior to both Truxima infusions to decrease the incidence and severity of infusion-related reactions (see Precautions).
First infusion: The recommended initial rate for infusion is 50 mg/hr; after the first 30 minutes, it can be escalated in 50 mg/hr increments every 30 minutes, to a maximum of 400 mg/hr.
Subsequent infusions: Subsequent doses of Truxima can be infused at an initial rate of 100 mg/hr, and increased by 100 mg/hr increments at 30 minutes intervals, to a maximum of 400 mg/hr.
Alternative subsequent, faster, infusion schedule: If patients did not experience a serious infusion-related reaction with their first or subsequent infusions of a dose of 1000 mg Truxima administered over the standard infusion schedule, a more rapid infusion can be administered for second and subsequent infusions using the same concentration as in previous infusions (4 mg/mL in a 250 mL volume). Initiate at a rate of 250 mg/hour for the first 30 minutes and then 600 mg/hour for the next 90 minutes. If the more rapid infusion is tolerated, this infusion schedule can be used when administering subsequent infusions.
Patients who have clinically significant cardiovascular disease, including arrhythmias, or previous serious infusion reactions to any prior biologic therapy or to rituximab, should not be administered the more rapid infusion.
Adult Patients with Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA): Induction of Remission: The recommended dosage of Truxima for induction of remission therapy in adult patients with GPA and MPA is 375 mg/m2 BSA, administered as an IV infusion once weekly for 4 weeks (four infusions in total).
Methylprednisolone given IV for 1 to 3 days and a dose of 1000 mg per day is recommended in combination with Truxima to treat severe vasculitis symptoms, followed by oral prednisone 1 mg/kg/day (not to exceed 80 mg/day, and tapered as rapidly as possible per clinical need) during and after the 4 week induction course of Truxima treatment.
Maintenance Treatment: Following induction of remission with Truxima, maintenance treatment in adult patients with GPA and MPA, should be initiated no sooner than 16 weeks after the last Truxima infusion.
Following induction of remission with other standard of care immunosuppressants, Truxima maintenance treatment should be initiated during the 4 week period that follows disease remission.
Administer Truxima as two 500 mg IV infusions separated by two weeks, followed by a 500 mg IV infusion at month 6, 12 and 18 and then every 6 months thereafter based on clinical evaluation. Patients should receive Truxima for at least 24 months after achievement of remission (absence of clinical signs and symptoms). For patients who may be at higher risk for relapse, physicians should consider a longer duration of Truxima maintenance therapy, up to 5 years.
First infusion: The recommended initial rate for infusion is 50 mg/hr; after the first 30 minutes, it can be escalated in 50 mg/hr increments every 30 minutes, to a maximum of 400 mg/hr.
Subsequent infusions: Subsequent doses of Truxima can be infused at an initial rate of 100 mg/hr, and increased by 100 mg/hr increments at 30 minutes intervals, to a maximum of 400 mg/hr.
Pediatric Patients with Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA): Induction of remission: The recommended dosage of Truxima for induction of remission therapy in pediatric patients with severe, active GPA/MPA is 375 mg/m2 BSA, administered as an IV infusion once weekly for 4 weeks.
Prior to the first Truxima infusion, methylprednisolone should be given IV for three daily doses of 30 mg/kg/day (not to exceed 1 g/day) to treat severe vasculitis symptoms. Up to three additional daily doses of 30 mg/kg IV methylprednisolone can be given prior to the first Truxima infusion.
Following completion of IV methylprednisolone, patients should receive oral prednisone 1 mg/kg/day (not to exceed 60 mg/day) and tapered as rapidly as possible per clinical need (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
Pemphigus Vulgaris: The recommended dosage of Truxima for the treatment of pemphigus vulgaris is 1000 mg administered as an IV infusion followed two weeks later by a second 1000 mg IV infusion in combination with a tapering course of glucocorticoids (see General as previously mentioned).
Maintenance Treatment: Maintenance infusions of 500 mg IV should be administered at month 12 and then every 6 months thereafter based on clinical evaluation.
Treatment of Relapse: In the event of relapse during the course of Truxima therapy, patients may receive 1000 mg IV. The healthcare provider should also consider resuming or increasing the patient's glucocorticoid dose based on clinical evaluation.
Subsequent infusions may be administered no sooner than 16 weeks following the previous infusion.
Special Dosage Instructions: Children and adolescents: No dose adjustments are recommended in pediatric patients (≥2 to <18 years of age) with active GPA/MPA. Currently available data are described in Use in Children under Precautions, PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies and Pharmacokinetics: Pharmacokinetics in Special Populations under Actions.
The safety and efficacy of Truxima in pediatric patients ≥ 6 months to < 18 years of age have not been established in indications other than previously untreated advanced stage CD20 positive DLBCL/BL/BAL/BLL. Only limited data are available for patients under 3 years of age (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
Truxima should not be used in pediatric patients from birth to < 6 months of age with CD20 positive diffuse large B-cell lymphoma (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
The safety and efficacy of Truxima in children and adolescents (≥2 to <18 years) have not been established in indications other than severe, active GPA or MPA.
Truxima should not be used in pediatric patients with severe, active GPA/MPA <2 years of age (see Use in Children under Precautions) as there is a possibility of an inadequate immune response towards childhood vaccinations against common, vaccine preventable childhood diseases (e.g. measles, mumps, rubella, and poliomyelitis) (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
Elderly: No dose adjustment is required in elderly patients (aged > 65 years).
Route of Administration: Intravenous (IV) infusion.
Overdosage
Limited experience with doses higher than the approved dose of intravenous rituximab formulation is available from clinical trials in humans. The highest intravenous dose of rituximab tested in humans to date is 5000 mg (2250 mg/m2), tested in a dose escalation study in patients with CLL. No additional safety signals were identified.
Patients who experience overdose should have immediate interruption of their infusion and be closely monitored.
In the post-marketing setting five cases of rituximab overdose have been reported. Three cases had no reported adverse event. The two adverse events that were reported were flu-like symptoms, with a dose of 1.8 g of rituximab and fatal respiratory failure, with a dose of 2 g of rituximab.
Contraindications
Contraindications for use in non-Hodgkin's lymphoma and chronic lymphocytic leukaemia: Hypersensitivity to the active substance or to murine proteins, or to any of the other excipients listed in Description.
Active, severe infections (see Precautions).
Patients in a severely immunocompromised state.
Contraindications for use in rheumatoid arthritis, granulomatosis with polyangiitis, microscopic polyangiitis and pemphigus vulgaris: Hypersensitivity to the active substance or to murine proteins, or to any other excipients listed in Description.
Active, severe infections (see Precautions).
Patients in a severely immunocompromised state.
Severe heart failure (New York Heart Association Class IV) or severe, uncontrolled cardiac disease (see Cardiovascular under Precautions).
Special Precautions
General: In order to improve the traceability of biological medicinal products, the trade name and batch number of the administered product should be clearly recorded (or stated) in the patient file.
Non-Hodgkin's Lymphoma and Chronic Lymphocytic Leukaemia Patients: Infusion-related reactions: Rituximab is associated with infusion-related reactions, which may be related to release of cytokines and/or other chemical mediators. Cytokine release syndrome may be clinically indistinguishable from acute hypersensitivity reactions.
Infusion-related reactions for Truxima: Severe infusion-related reactions with fatal outcome have been reported during post-marketing use. Severe infusion-related reactions usually manifested within 30 minutes to 2 hours after starting the first rituximab infusion, were characterized by pulmonary events and included, in some cases, rapid tumour lysis and features of tumour lysis syndrome in addition to fever, chills, rigors, hypotension, urticaria, angioedema and other symptoms (see Adverse Reactions). Patients with a high tumour burden or with a high number (> 25 x 109/L) of circulating malignant cells such as patients with CLL and mantle cell lymphoma may be at higher risk of developing severe infusion-related reactions. Infusion reaction symptoms are usually reversible with interruption of the infusion. Treatment of infusion-related symptoms with diphenhydramine and acetaminophen is recommended. Additional treatment with bronchodilators or IV saline may be indicated. In most cases, the infusion can be resumed at a 50% reduction in rate (e.g. from 100 mg/h to 50 mg/h) when symptoms have completely resolved. Most patients who have experienced non-life threatening infusion-related reactions have been able to complete the full course of rituximab therapy. Further treatment of patients after complete resolution of signs and symptoms has rarely resulted in repeated severe infusion-related reactions. Anaphylactic and other hypersensitivity reactions have been reported following the intravenous administration of proteins to patients. Epinephrine, antihistamines and glucocorticoids should be available for immediate use in the event of a hypersensitivity reaction to rituximab.
Patients with a high number [> 25 x 109/L] of circulating malignant cells or high tumour burden such as patients with CLL and mantle cell lymphoma, who may be at higher risk of especially severe infusion-related reactions, should only be treated with extreme caution. These patients should be very closely monitored throughout the first infusion. Consideration should be given to the use of a reduced infusion rate for the first infusion in these patients or a split dosing over two days during the first cycle and any subsequent cycles if the lymphocyte count is still > 25 x 109/L.
Hypersensitivity reactions/Anaphylaxis: Anaphylactic and other hypersensitivity reactions have been reported following the intravenous administration of proteins to patients. Epinephrine, antihistamines and glucocorticoids should be available for immediate use in the event of a hypersensitivity reactions to rituximab.
Pulmonary events: Pulmonary events have included hypoxia, pulmonary infiltration, and acute respiratory failure. Some of these events have been preceded by severe bronchospasm and dyspnea. In some cases, symptoms worsened over time, while in others initial improvement was followed by clinical deterioration. Therefore, patients experiencing pulmonary events or other severe infusion-related symptoms should be closely monitored until complete resolution of their symptoms occurs. Patients with a history of pulmonary insufficiency or those with pulmonary tumour infiltration may be at greater risk of poor outcome and should be treated with increased caution. Acute respiratory failure may be accompanied by events such as pulmonary interstitial infiltration or edema, visible on a chest x-ray. The syndrome usually manifests itself within one or two hours of initiating the first infusion. Patients who experience severe pulmonary events should have their infusion interrupted immediately (see Dosage & Administration) and should receive aggressive symptomatic treatment.
Rapid tumour lysis: Rituximab mediates the rapid lysis of benign and malignant CD20-positive cells. Signs and symptoms (e.g. hyperuricemia, hyperkalemia, hypocalcaemia, hyperphosphataemia, acute renal failure, elevated LDH) consistent with tumour lysis syndrome (TLS) have been reported to occur after the first rituximab infusion in patients with high numbers of circulating malignant lymphocytes. Prophylaxis for TLS should be considered for patients at risk of developing rapid tumour lysis (e.g. patients with a high tumour burden or with a high number (> 25 x 109/L) of circulating malignant cells such as patients with CLL and mantle cell lymphoma). These patients should be followed closely and appropriate laboratory monitoring performed. Appropriate medical therapy should be provided for patients who develop signs and symptoms consistent with rapid tumour lysis. Following treatment for and complete resolution of signs and symptoms, subsequent rituximab therapy has been administered in conjunction with prophylactic therapy for TLS in a limited number of cases.
Cardiovascular: Since hypotension may occur during rituximab infusion, consideration should be given to withholding antihypertensive medications 12 hours prior to and throughout rituximab infusion. Angina pectoris or cardiac arrhythmia, such as atrial flutter and fibrillation, have occurred in patients treated with rituximab. Therefore patients with a history of cardiac disease should be monitored closely.
Monitoring of blood counts: Although rituximab is not myelosuppressive in monotherapy, caution should be exercised when considering treatment of patients with neutrophil counts of < 1.5 x 109/L and/or platelet counts of < 75 x 109/L, as clinical experience with such patients is limited. Rituximab has been used in patients who underwent autologous bone marrow transplantation and in other risk groups with a presumable reduced bone marrow function without inducing myelotoxicity. Consideration should be given to the need for regular full blood counts, including platelet counts, during monotherapy with rituximab. When rituximab is given in combination with CHOP or CVP chemotherapy, regular full blood counts should be performed according to usual medical practice.
Infections: Rituximab treatment should not be initiated in patients with severe active infections.
Hepatitis B Infections: Cases of hepatitis B reactivation, including reports of fulminant hepatitis, some of which were fatal, have been reported in subjects receiving rituximab, although the majority of these subjects were also exposed to cytotoxic chemotherapy. The reports are confounded by both the underlying disease state and the cytotoxic chemotherapy. Hepatitis B virus (HBV) screening should be performed in all patients before initiation of treatment with rituximab. At minimum this should include HBsAg-status and HBcAb-status. These can be complemented with other appropriate markers as per local guidelines. Patients with active hepatitis B should be not be treated with rituximab. Patients with positive hepatitis B serology should consult liver disease experts before start of treatment and should be monitoring and managed following local medical standards to prevent hepatitis B reactivation.
Progressive multifocal leukoencephalopathy (PML): Cases of progressive multifocal leukoencephalopathy (PML) have been reported during use of rituximab in NHL and CLL (see Adverse Reactions and Post Marketing under Adverse Reactions). The majority of patients had received rituximab in combination with chemotherapy or as part of a haematopoietic stem cell transplant. Physicians treating patients with NHL or CLL should consider PML in the differential diagnosis of patients reporting neurological symptoms and consultation with a neurologist should be considered as clinically indicated.
Skin reactions: Severe skin reactions such as Toxic Epidermal Necrolysis (Lyell's syndrome) and Stevens-Johnson syndrome, some with fatal outcome, have been reported (see Post Marketing under Adverse Reactions). In case of such an event, with a suspected relationship to rituximab, treatment should be permanently discontinued.
Immunization: The safety of immunization with live viral vaccines, following rituximab therapy has not been studied and vaccination with live virus vaccines is not recommended.
Patients treated with rituximab may receive non-live vaccinations. However, with non-live vaccines response rates may be reduced. In a non-randomized study, patients with relapsed low-grade NHL who received rituximab monotherapy when compared to healthy untreated controls had a lower rate of response to vaccination with tetanus recall antigen (16% Vs 81%) and Keyhole Limpet Haemocyanin (KLH) neoantigen (4% Vs 69% when assessed for > 2-fold increase in antibody titer).
Mean pre-therapeutic antibody titers against a panel of antigens (Streptococcus pneumoniae, influenza A, mumps, rubella, varicella) were maintained for at least 6 months after treatment with rituximab.
Rheumatoid Arthritis Patients (RA), Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA) Patients and Pemphigus Vulgaris (PV) Patients: The efficacy and safety of rituximab for the treatment of autoimmune diseases other than rheumatoid arthritis, granulomatosis with polyangiitis (Wegener's) and microscopic polyangiitis and pemphigus vulgaris have not been established.
Infusion-related Reactions: Rituximab is associated with infusion-related reactions (IRRs), which may be related to release of cytokines and/or other chemical mediators. For RA patients, most infusion-related events reported in clinical trials were mild to moderate in severity. Severe IRRs with fatal outcome have been reported in the post-marketing setting (see Post Marketing under Adverse Reactions). Closely monitor patients with pre-existing cardiac conditions and those who experienced prior cardiopulmonary adverse reactions. The most common symptoms were headache, pruritus, throat irritation, flushing, rash, urticaria, hypertension, and pyrexia. In general, the proportion of patients experiencing any infusion reaction was higher following the first infusion of any treatment course than following the second infusion. Subsequent rituximab infusions were better tolerated by patients than the initial infusion. Less than 1% of patients experienced serious IRRs, with most of these reported during the first infusion of the first course (see Adverse Reactions). The reactions reported were usually reversible with a reduction in rate or interruption of rituximab infusion, and administration of an anti-pyretic, an antihistamine and occasionally oxygen, IV saline, bronchodilators, or glucocorticoids as required. In most cases, the infusion can be resumed at a 50% reduction in rate (e.g. from 100 mg/h to 50 mg/h) when symptoms have completely resolved.
Infusion-related reactions for GPA/MPA and PV patients were consistent with those seen for RA patients in clinical trial (see Adverse Reactions).
Hypersensitivity Reactions/Anaphylaxis: Anaphylactic and other hypersensitivity reactions have been reported following the IV administration of proteins to patients. Medicinal products for the treatment of hypersensitivity reactions (e.g. epinephrine, antihistamines and glucocorticoids), should be available for immediate use in the event of an allergic reaction during administration of rituximab.
Cardiovascular: Since hypotension may occur during rituximab infusion, consideration should be given to withholding anti-hypertensive medications 12 hours prior to the rituximab infusion.
Angina pectoris, cardiac arrhythmias such as atrial flutter and fibrillation, heart failure or myocardial infarction have occurred in patients treated with rituximab. Therefore patients with a history of cardiac disease should be monitored closely (see Infusion-related Reactions as previously mentioned).
Infections: Based on the mechanism of action of rituximab and the knowledge that B cells play an important role in maintaining normal immune response, patients may have an increased risk of infection following rituximab therapy. Rituximab should not be administered to patients with an active infection or severely immunocompromised patients (e.g. where levels of CD4 or CD8 are very low). Physicians should exercise caution when considering the use of rituximab in patients with a history of recurring or chronic infections or with underlying conditions which may further predispose patients to serious infection (see Adverse Reactions). Patients who develop infection following rituximab therapy should be promptly evaluated and treated appropriately.
Hepatitis B Infections: Cases of hepatitis B reactivation including those with a fatal outcome have been reported in RA, GPA and MPA patients receiving rituximab. Hepatitis B virus (HBV) screening should be performed in all patients before initiation of treatment with rituximab. At minimum this should include HBsAg-status and HBcAb-status. These can be complemented with other appropriate markers as per local guidelines. Patients with active hepatitis B disease should not be treated with rituximab. Patients with positive hepatitis B serology should consult liver disease experts before start of treatment and should be monitored and managed following local medical standards to prevent hepatitis B reactivation.
Skin reactions: Severe skin reactions such as toxic epidermal necrolysis and Stevens-Johnson syndrome, some with fatal outcome, have been reported (see Post Marketing under Adverse Reactions). In case of such an event with a suspected relationship to rituximab, treatment should be permanently discontinued.
Progressive Multifocal Leukoencephalopathy (PML): Cases of fatal progressive multifocal leukoencephalopathy have been reported following use of rituximab for the treatment of autoimmune diseases including RA. Several, but not all of the reported cases had potential risk factors for PML, including the underlying disease, long-term immunosuppressive therapy or chemotherapy. PML has also been reported in patients with autoimmune disease not treated with rituximab. Physicians treating patients with autoimmune diseases should consider PML in the differential diagnosis of patients reporting neurological symptoms and consultation with a neurologist should be considered as clinically indicated.
Immunization: The safety of immunization with live viral vaccines following rituximab therapy has not been studied. Therefore vaccination with live virus vaccines is not recommended whilst on rituximab or whilst peripherally B cell depleted. Patients treated with rituximab a may receive non-live vaccinations. However, response rates to non-live vaccines may be reduced.
For patients, treated with rituximab, physicians should review the patient's vaccination status and patients should, if possible, be brought up-to-date with all immunizations in agreement with current immunization guidelines prior to initiating rituximab therapy. Vaccination should be completed at least 4 weeks prior to first administration of rituximab.
In a randomized study, patients with RA treated with rituximab and methotrexate had comparable response rates to tetanus recall antigen (39% Vs 42%), reduced rates to pneumococcal polysaccharide vaccine (43% Vs 82% to at least 2 pneumococcal antibody serotypes), and KLH neoantigen (34% Vs 80%), when given at least 6 months after rituximab as compared to patients only receiving methotrexate. Should non-live vaccinations be required whilst receiving rituximab therapy, these should be completed at least 4 weeks prior to commencing the next course of rituximab.
In the overall experience of rituximab repeat treatment over one year, the proportions of patients with positive antibody titers against S. pneumoniae, influenza, mumps, rubella, varicella and tetanus toxoid were generally similar to the proportions at baseline.
Methotrexate (MTX) naïve RA populations: The use of rituximab is not recommended in MTX-naïve patients since a favourable benefit risk relationship has not been established.
DRUG ABUSE AND DEPENDENCE: No data to report.
Ability to Drive and Use Machines: Rituximab has no or negligible effect on the ability to drive and use machines.
Renal Impairment: The safety and efficacy of renal impairment in rituximab patients has not been established.
Hepatic Impairment: The safety and efficacy of hepatic impairment in rituximab patients has not been established.
Use in Children: Only limited data are available for patients under 3 years of age. See PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions.
Pediatric Patients with Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA): In a study, twenty-five pediatric patients (6 children ≥ 2 years to < 12 years and 19 adolescents ≥ 12 years to < 18 years) with active GPA/MPA were administered rituximab (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
In general, the adverse drug reactions observed in rituximab treated pediatric patients with active GPA/MPA during the overall study period were consistent in type, nature and severity to those seen in adult patients (see Clinical Trials under Adverse Reactions).
The efficacy of rituximab in pediatric active GPA/MPA patients is based on PK exposure from WA25615 (PePRS) and extrapolation from the established efficacy of rituximab in adult GPA/MPA patients (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies and Pharmacokinetics: Pharmacokinetics in Special Populations under Actions).
The safety and efficacy of rituximab in pediatric patients has not been studied in disease other than GPA/MPA. Rituximab should not be used in pediatric patients with GPA/MPA < 2 years of age as there is a possibility of an inadequate immune response towards childhood vaccination against common, vaccine preventable childhood disease (e.g. measles, mumps, rubella, and poliomyelitis).
Hypogammaglobulinaemia has been observed in pediatric patients treated with rituximab, in some cases severe and requiring long-term immunoglobulin substitution therapy (see Clinical Trials under Adverse Reactions). The consequences of long term B-cell depletion in pediatric patients are unknown.
Use in the Elderly: The safety and efficacy of rituximab in geriatric patients has been established.
Use In Pregnancy & Lactation
Females and Males of Reproductive Potential: Fertility: No preclinical fertility studies have been conducted.
Animal data: Developmental toxicity studies performed in cynomolgus monkeys revealed no evidence of embryotoxicity in utero. Newborn offspring of maternal animals exposed to rituximab were noted to have depleted B cell populations during the post natal phase.
Contraception: Women of childbearing age should employ effective contraceptive methods during and for up to 12 months after treatment with rituximab.
Pregnancy: IgG immunoglobulins are known to cross the placental barrier.
B cell levels in human neonates following maternal exposure to rituximab have not been studied in clinical trials. There are no adequate and well-controlled data from studies in pregnant women, however transient B-cell depletion and lymphocytopenia have been reported in some infants born to mothers exposed to rituximab during pregnancy. For these reasons rituximab should not be administered to pregnant women unless the possible benefit outweighs the potential risk.
Nursing Mothers: It is not known whether rituximab is excreted in human breast milk. Given, however, that maternal IgG enters breast milk, rituximab should not be administered to nursing mothers.
Adverse Reactions
Clinical Trials: Experience from Clinical Trials in Haemato-Oncology: The frequencies of adverse drug reactions (ADRs) reported with rituximab alone or in combination with chemotherapy are summarized in the tables as follows and are based on data from clinical trials. These ADRs had either occurred in single arm studies or had occurred with at least a 2% difference compared to the control arm in at least one of the major randomized clinical trials. ADRs are added to the appropriate category in the tables as follows according to the highest incidence seen in any of the major clinical trials. Within each frequency grouping ADRs are listed in descending order of severity. Frequencies are defined as very common (≥ 1/10), common (≥ 1/100 to < 1/10), and uncommon (≥ 1/1,000 to < 1/100).
Rituximab monotherapy/maintenance therapy: The ADRs in Table 25 are based on data from single-arm studies including 356 patients with low-grade or follicular lymphoma, treated with rituximab weekly as single agent for the treatment or re-treatment of Non-Hodgkin's Lymphoma up to 4 weeks in most patients and from 25 patients who received doses other than 375 mg/m2 for four doses and up to 500 mg/m2 single dose in the Phase I setting (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions for further details). The table also contains ADRs based on data from 671 patients with follicular lymphoma who received rituximab as maintenance therapy for up to 2 years following response to initial induction with CHOP, R-CHOP, R-CVP or R-FCM (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions for further details). The ADRs were reported up to 12 months after treatment with monotherapy and up to 1 month after treatment with rituximab maintenance. (See Table 25.)

Click on icon to see table/diagram/image

Rituximab in combination with chemotherapy in NHL and CLL: The ADRs listed in Table 26 are based on rituximab-arm data from controlled clinical trials that occurred in addition to those seen with monotherapy/maintenance therapy and/or at a higher frequency grouping: 202 patients with diffuse large B-cell lymphoma (DLBCL) treated with R-CHOP, and from 234 and 162 patients with follicular lymphoma treated with R-CHOP or R-CVP, respectively and from 397 previously untreated CLL patients and 274 relapsed/refractory CLL patients, treated with rituximab in combination with fludarabine and cyclophosphamide (R-FC) (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions for further details). (See Table 26.)

Click on icon to see table/diagram/image

The following terms have been reported as adverse events, however, were reported at a similar (< 2% difference between the groups) or lower incidence in the rituximab-arms compared to control arms: Haematotoxicity, neutropenic infection, urinary tract infection, septic shock, superinfection lung, implant infection, septicaemia staphylococcal, lung infection, rhinorrheoa, pulmonary oedema, cardiac failure, sensory disturbance, venous thrombosis, mucosal inflammation nos, influenza-like illness, oedema lower limb, abnormal ejection fraction, pyrexia, general physical health deterioration, fall, multi-organ failure, venous thrombosis deep limb, positive blood culture, diabetes mellitus inadequate control.
The safety profile for rituximab in combination with other chemotherapies (e.g. MCP, CHVP-IFN) is comparable to the safety profile as described for the combination of rituximab and CVP, CHOP or FC in equivalent populations.
Further information on selected, serious adverse drug reactions: Administration-related reactions: Monotherapy 4 weeks treatment: Signs and symptoms suggestive of an infusion-related reaction (IRR) were reported in more than 50% of patients in clinical trials, and were predominantly seen during the first infusion. Hypotension, fever, chills, rigors, urticarial, bronchospasm, sensation of tongue or throat swelling (angioedema), nausea, fatigue, headache, pruritus, dyspnea, rhinitis, vomiting, flushing, and pain at disease sites have occurred in associated with rituximab infusion as part of an infusion-related symptom complex. Some features of tumor lysis syndrome have also been observed.
Combination Therapy (R-CVP in NHL; R-CHOP in DLBCL, R-FC in CLL): Severe IRRs occurred in up to 12% of all patients at the time on the first treatment cycle with rituximab in combination with chemotherapy. The incidence of infusion-related symptoms decreased substantially with subsequent infusions and in <1% of patients by the eighth cycle. Additional reactions reported were dyspepsia, rash, hypertension, tachycardia, and features of tumour lysis syndrome. Isolated cases of myocardial infarction, atrial fibrillation, pulmonary oedema and acute reversible thrombocytopenia were also reported.
Infusion-related reactions: Monotherapy - 4 weeks treatment: Hypotension, fever, chills, rigors, urticaria, bronchospasm, sensation of tongue or throat swelling (angioedema), nausea, fatigue, headache, pruritus, dyspnea, rhinitis, vomiting, flushing, and pain at disease sites have occurred in association with rituximab infusion as part of an infusion-related symptom complex. Such infusion-related symptoms occurred in the majority of patients during the first rituximab infusion (see General under Precautions). The incidence of infusion-related symptoms decreased from 77% (7% grade 3/4) with the first infusion to approximately 30% (2% grade 3/4) with the fourth infusion and to 14% (no grade 3/4 events) with the eighth infusion.
Maintenance Treatment (NHL) up to 2 years: Non-serious signs and symptoms suggestive of an infusion-related reaction were reported in 41% of patients under general disorders (mainly asthenia, pyrexia, influenza like illness, pain) and in 7% of patients for immune system disorders (hypersensitivity). Serious infusion-related reactions occurred in <1% of patients.
Combination Therapy (R-CVP in NHL; R-CHOP in DLBCL, R-FC in CLL): Severe infusion-related reactions occurred in up to 12% of all patients at the time of the first treatment cycle with rituximab in combination with chemotherapy. The incidence of severe infusion-related reactions decreased to less than 1% by the eighth cycle of therapy. The signs and symptoms were consistent with those observed during monotherapy (see Precautions and Experience from Clinical Trials in Haemato-Oncology: Rituximab monotherapy/maintenance therapy as previously mentioned), but also included dyspepsia, rash, hypertension, tachycardia, features of tumour lysis syndrome. Additional reactions reported in isolated cases at the time of R-chemotherapy were myocardial infarction, atrial fibrillation, pulmonary oedema and acute reversible thrombocytopenia.
Infections: Monotherapy - 4 weeks treatment: Rituximab induced B-cell depletion in 70% to 80% of patients but was associated with decreased serum immunoglobulins in only a minority of patients. Bacterial, viral, fungal and unknown etiology infections, irrespective of causal assessment, occurred in 30.3% of 356 patients: Severe infectious events (grade 3 or 4), including sepsis occurred in 3.9% of patients.
Maintenance Treatment (NHL) up to 2 years: Higher frequencies of infections overall, including Grade 3 and 4 infections, were observed during rituximab treatment. There was no cumulative toxicity in terms of infections reported over the 2-year maintenance period. Data from clinical trial included two cases of fatal PML in NHL patients that occurred after disease progression and retreatment (see Precautions).
Combination Therapy (R-CVP in NHL; R-CHOP in DLBCL, R-FC in CLL): No increase in the frequency of infections or infestations was observed. The most common infections were upper respiratory tract infections which were reported for 12.3% patients on R-CVP and 16.4% patients receiving CVP; No life-threatening infections were reported during this study.
In the R-CHOP study the overall incidence of grade 2 to 4 infections was 45.5% in the R-CHOP group and 42.3% in the CHOP group. Grade 2 to 4 fungal infections were more frequent in the R-CHOP group (4.5% vs 2.6% in the CHOP group); this difference was due to a higher incidence of localized Candida infections during the treatment period. The incidence of grade 2 to 4 herpes zoster was higher in the R-CHOP group (4.5%) than in the CHOP group (1.5%). The proportion of patients with grade 2 to 4 infections and/or febrile neutropenia was 55.4% in the R-CHOP group and 51.5% in the CHOP group. Febrile neutropenia (i.e. no report of concomitant documented infection) was reported only during the treatment period, in 20.8% in the R-CHOP group and 15.3% in the CHOP group.
In patients with CLL, the incidence of grade 3 or 4 hepatitis B infection (reactivation and primary infection) was 2% R-FC vs 0% in the FC group.
Hematologic events: Monotherapy - 4 weeks: Severe (grade 3 and 4) neutropenia was reported in 4.2% of patients, severe anaemia was reported in 1.1% of patients and severe thrombocytopenia was reported in 1.7% of patients.
Maintenance Treatment (NHL) up to 2 years: There was a higher incidence of grade 3-4 leucopenia (observation 2%, rituximab 5%) and neutropenia (observation 4%, rituximab 10%) in the rituximab arm compared to the observation arm. The incidence of grade 3 to 4 thrombocytopenia (observation 1%, rituximab <1%) was low. In approximately half of the patients with available data on B-cell recovery after end of rituximab induction treatment, it took 12 months or more for their B-cell levels to return to normal values.
Combination Therapy (R-CVP in NHL; R-CHOP in DLBCL, R-FC in CLL): During treatment course in studies with rituximab in combination with chemotherapy, Grade 3 and 4 leucopenia (R-CHOP 88% vs. CHOP 79%, R-FC 23% vs. FC 12%) and neutropenia (R-CVP 24% vs. CVP 14%; R-CHOP were usually reported with higher frequencies when compared to chemotherapy alone). However, the higher incidence of neutropenia in patients treated with rituximab and chemotherapy was not associated with a higher incidence of infections and infestations compared to patients treated with chemotherapy alone. Studies in previously untreated and relapsed/refractory CLL have established that in some cases neutropenia was prolonged or with a late onset following treatment in rituximab plus FC group. No relevant difference between the treatment arms was observed with respect to Grade 3 and 4 anaemia or thrombocytopenia. In CLL first line, Grade 3 and 4 anaemia was reported by 4% of patients treated with R-FC compared to 7% of patients receiving FC, and Grade 3 and 4 thrombocytopenia was reported by 7% of patients in the R-FC group compared to 10% of patients in the FC group. In the relapsed/refractory CLL study, adverse events of Grade 3 and 4 anaemia were reported in 12% of patients treated with R-FC compared to 13% of patients receiving FC and Grade 3 and 4 thrombocytopenia was reported by 11% of patients in the R-FC group compared to 9% of patients in the FC group.
Cardiovascular events: Monotherapy - 4 weeks treatment: Cardiovascular events were reported in 18.8% of patients during the treatment period. The most frequently reported events were hypotension and hypertension. Cases of Grade 3 and 4 arrhythmia (including ventricular and supraventricular tachycardia) and angina pectoris during a rituximab infusion were reported.
Maintenance Treatment (NHL) up to 2 years: The incidence of grade 3 to 4 cardiac disorders was comparable between the two treatment groups. Cardiac events were reported as serious adverse event in < 1% of patients on observation and in 3% of patients on rituximab: atrial fibrillation (1%), myocardial infarction (1%), left ventricular failure (< 1%), myocardial ischemia (< 1%).
Combination Therapy (R-CVP in NHL; R-CHOP in DLBCL, R-FC in CLL): In the R-CHOP study the incidence of grade 3 and 4 cardiac arrhythmias, predominantly supraventricular arrhythmias such as tachycardia and atrial flutter/fibrillation, was higher in the R-CHOP group (6.9%) as compared to the CHOP group (1.5%). All of these arrhythmias either occurred in the context of a rituximab infusion or were associated with predisposing conditions such as fever, infection, acute myocardial infarction or pre-existing respiratory and cardiovascular disease (see Precautions). No difference between the R-CHOP and CHOP group was observed in the incidence of other grade 3 and 4 cardiac events including heart failure, myocardial disease and manifestations of coronary artery disease.
In CLL, the overall incidence of grade 3 or 4 cardiac disorders was low both in the first-line study (4% R-FC, 3% FC) and in the relapsed/refractory study (4% R-FC, 4% FC).
IgG levels: Maintenance Treatment (NHL) up to 2 years: After induction treatment, median IgG levels were below the lower limit of normal (LLN) (< 7 g/L) in both the observation and the rituximab groups. In the observation group, the median IgG level subsequently increased to above the LLN, but remained constant during rituximab treatment. The proportion of patients with IgG levels below the LLN was about 60% in the rituximab group throughout the 2 year treatment period, while it decreased in the observation group (36% after 2 years).
A small number of spontaneous and literature cases of hypogammaglobulinaemia have been observed in pediatric patients treated with rituximab, in some cases severe and requiring long-term immunoglobulin substitution therapy. The consequences of long-term B cell depletion in pediatric patients are unknown.
Neurologic events: Combination Therapy (R-CVP in NHL; R-CHOP in DLBCL, R-FC in CLL): During the treatment period, (2% of patients) in the R-CHOP group, all with cardiovascular risk factors, experienced thromboembolic cerebrovascular accidents during the first treatment cycle. There was no difference between the treatment groups in the incidence of other thromboembolic events. In contrast, three patients (1.5%) had cerebrovascular events in the CHOP group, all of which occurred during the follow-up period.
In CLL, the overall incidence of grade 3 or 4 nervous system disorders was low both in the first-line study (4% R-FC, 4% FC) and in the relapsed/refractory study (3% R-FC, 3% FC).
Subpopulations: Monotherapy - 4 weeks treatment: Elderly patients (≥ 65 years): The incidence of any ADR and of grade 3 and 4 ADRs was similar in elderly (n = 94) and younger (n = 237) patients (88.3% versus 92.0% for any ADR and 16.0% versus 18.1% for grade 3 and 4 ADRs).
Combination Therapy: Elderly patients (≥ 65 years): The incidence of grade 3/4 blood and lymphatic adverse events was higher in elderly patients (≥ 65 years of age) compared to younger patients, with previously untreated or relapsed/refractory CLL.
Bulky disease: Patients with bulky disease (n = 39) had a higher incidence of grade 3 and 4 ADRs than patients without bulky disease (n = 195; 25.6% versus 15.4%). The incidence of any ADR was similar in these two groups (92.3% in bulky disease versus 89.2% in non-bulky disease).
Re-treatment with Monotherapy: The percentage of patients reporting any ADR and grade 3 and 4 ADRs upon re-treatment with further courses of rituximab was similar to the percentage of patients reporting any ADR and grade 3 and 4 ADRs upon initial exposure (95.0% versus 89.7% for any ADR and 13.3% versus 14.8% for grade 3 and 4 ADRs).
Experience from pediatric DLBCL/BL/BAL/BLL Clinical Trials: Summary of safety profile: A multicenter, open-label randomized study of Lymphome Malin B chemotherapy (LMB) with or without rituximab was conducted in pediatric patients (aged ≥ 6 months to < 18 years old) with previously untreated advanced stage CD20 positive DLBCL/BL/BAL/BLL.
A total of 309 pediatric patients received rituximab and were included in the safety analysis population. Pediatric patients randomized to the LMB chemotherapy arm with rituximab, or enrolled in the single arm part of the study, were administered rituximab at a dose of 375 mg/m2 BSA and received a total of six intravenous infusions of rituximab (two during each of the two induction courses and one during each of the two consolidation courses of the LMB scheme).
The safety profile of rituximab in pediatric patients (aged ≥ 6 months to < 18 years old) with previously untreated advanced stage CD20 positive DLBCL/BL/BAL/BLL was generally consistent in type, nature and severity with the known safety profile in adult NHL and CLL patients. Addition of rituximab to chemotherapy did result in an increased risk of some events including infections (including sepsis) compared to chemotherapy only.
Experience from Rheumatoid Arthritis Clinical Trials: The safety profile of rituximab in the treatment of patients with moderate to severe RA is summarized in the sections as follows. In the all exposure population more than 3000 patients have received at least one treatment course and were followed for periods ranging from 6 months to over 5 years with an overall exposure equivalent to 7198 patient years; approximately 2300 patients received two or more courses of treatment during the follow-up period.
The ADRs listed in Table 27 are based on data from placebo-controlled periods of four multicenter, RA clinical trials. The patient populations receiving rituximab differed between studies, ranging from early active RA patients who were methotrexate (MTX) naïve, through MTX inadequate responders (MTX-IR) to patients who had inadequate response to anti-TNF therapies (TNF-IR) (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions for further information).
Patients received either 2 x 1000 mg or 2 x 500 mg of rituximab separated by an interval of two weeks; in addition to methotrexate (10 - 25 mg/week) (see Rheumatoid arthritis (RA) under Dosage & Administration). The ADRs listed in Table 27 are those which occurred at a rate of at least 2%, with at least a 2% difference compared to the control arm and are presented regardless of dose. Frequencies in Table 25 and corresponding footnote are defined as very common (≥ 1/10), common (≥ 1/100 to < 1/10) and uncommon (≥ 1/1,000 to < 1/100). (See Table 27.)

Click on icon to see table/diagram/image

In the all exposure population, the safety profile was consistent with that seen in the controlled period of the clinical trials with no new ADRs identified.
Multiple Courses: Multiple courses of treatment are associated with a similar ADR profile to that observed following first exposure. The safety profile improved with subsequent courses due to a decrease in IRRs, RA exacerbation and infections, all of which were more frequent in the first 6 months of treatment.
Further information on selected adverse drug reactions: Infusion-related reactions: The most frequent ADRs following receipt of rituximab in RA clinical studies were IRRs. Among the 3095 patients treated with rituximab, 1077 (35%) experienced at least one IRR. The vast majority of IRRs were CTC Grade 1 or 2. In clinical studies less than 1% (14/3095 patients) of patients with RA who received an infusion of rituximab at any dose experienced a serious IRR. There were no CTC Grade 4 IRRs and no deaths due to IRRs. The proportion of CTC Grade 3 events, and of IRRs leading to withdrawal decreased by course and were rare from course 3 onwards. Signs and or symptoms suggesting an infusion-related reaction (nausea, pruritus, fever, urticaria/rash, chills, pyrexia, rigors, sneezing, angioneurotic oedema, throat irritation, cough and bronchospasm, with or without associated hypotension or hypertension) were observed in 720/3095 (23%) patients following first infusion of the first exposure to rituximab.
Premedication with IV glucocorticoid significantly reduced the incidence and severity of these events (see General: Rheumatoid Arthritis Patients (RA), Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA) Patients and Pemphigus Vulgaris (PV) Patients under Precautions).
Infections: The overall rate of infection was approximately 97 per 100 patient years in rituximab treated patients. The infections were predominately mild to moderate and consisted mostly of upper respiratory tract infections and urinary tract infections. The rate of serious infections was approximately 4 per 100 patient years, some of which were fatal. In addition to the ADRs in Table 25, medically serious events reported also include pneumonia at a frequency of 1.9%.
Malignancies: The incidence of malignancy following exposure to rituximab in clinical studies (0.8 per 100 patient years) lies within the range expected for an age and gender matched population.
Clinical Trial Experience in Adult and Pediatric Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA): Adult Induction of Remission (GPA/MPA Study 1): Ninety-nine adult GPA/MPA patients were treated for induction of remission of GPA and MPA in a clinical trial with rituximab (375 mg/m2, once weekly for 4 weeks) and glucocorticoids (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
The ADRs listed in Table 26 were all adverse events which occurred at an incidence of ≥10% in the rituximab treated group. Frequencies in Table 28 are defined as very common (≥1/10). (See Table 28.)

Click on icon to see table/diagram/image

Adult Maintenance Treatment (GPA/MPA Study 2): In a further clinical study, a total of 57 adult patient with severe active GPA and MPA were treated for the maintenance of remission (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
No new safety concerns were identified and the safety profile was consistent with the well-established safety profile for rituximab in approved autoimmune indications, including GPA/MPA. Overall, 4% of patients in the rituximab arm experienced adverse events leading to discontinuation.
Most adverse events in the rituximab arm were mild or moderate in intensity. No patients in the rituximab arm had fatal adverse events. ADRs were all adverse events which occurred at an incidence of ≥10% in the rituximab treated group. The very commonly (≥10%) reported events considered ADRs were: infusion-related reactions and infections.
Adult Long-term Follow-up (GPA/MPA Study 3): In a long-term observational safety study, 97 adult GPA/MPA patients received treatment with rituximab (mean of 8 infusions [range 1-28]) for up to 4 years, according to their physician's standard practice and discretion.
The overall safety profile was consistent with the well-established safety profile of rituximab in RA and GPA/MPA and no new adverse drug reactions were reported.
Pediatric Population: An open-label, single arm study was conducted in 25 pediatric patients with active GPA/MPA. The overall study period consisted of a 6-month remission induction phase and a minimum 18-month follow-up phase, up to 4.5 yrs. During the follow-up phase, rituximab was given at the discretion of the investigator (17 out of 25 patients received additional rituximab treatment). Concomitant treatment with other immunosuppressive therapy was permitted (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
All identified ADRs were considered all adverse events that occurred at an incidence of ≥10%. These included: infections (17 patients [68%] in the remission induction phase; 23 patients [92%] in the overall study period), IRRs (15 patients [60%] in the remission induction phase; 17 patients [68%] in the overall study period), and nausea (4 patients [16%] in the remission induction phase; 5 patients [20%] in the overall study period).
During the overall study period, the safety profile of rituximab in pediatric GPA/MPA patients was consistent in type, nature and severity with the known safety profile in adult patients for autoimmune disease, including adult GPA/MPA.
Further information on selected adverse drug reactions: Infusion-related reactions: In the clinical trial studying induction of remission (GPA/MPA study 1) in adult patients with severe active GPA and MPA infusion-related reactions (IRRs) were defined as any adverse event occurring within 24 hours of an infusion and considered to be infusion-related by investigators in the safety population. Of the 99 patients treated with rituximab 12/99 patients (12%) experienced at least one IRR. All IRRs were CTC Grade 1 or 2. The most common IRRs included cytokine release syndrome, flushing, throat irritation, and tremor.
Rituximab was given in combination with intravenous glucocorticoids which may reduce the incidence and severity of these events.
In the maintenance therapy clinical trial (GPA/MPA study 2) in adult patients, 7/57 (12%) patients in the rituximab arm reported infusion-related reactions. The incidence of IRR symptoms was highest during or after the first infusion (9%) and decrease with subsequent infusion (<4%). All IRR symptoms were mild to moderate and most were reported from the Respiratory, Thoracic and Mediastinal Disorders and Skin and Subcutaneous Tissue disorder SOCs.
In the clinical trial in pediatric patients with GPA/MPA, the reported IRRs were predominantly seen with the first infusion (8 patients [32%]), and then decreased over time with the number of rituximab infusions (20% with the second infusion, 12% with the third infusion and 8% with the fourth Infusion). The most common IRR symptoms reported during the remission of induction phase were headache, rash, rhinorrhea and pyrexia (8% for each symptom). The observed symptoms of IRRs were similar to those known in adult GPA/MPA patients treated with rituximab. The majority of IRRs were Grade 1 and Grade 2, there were two non-serious Grade 3 IRRs, and no Grade 4-5 IRRs reported. One serious Grade 2 IRR (generalized oedema which resolved with treatment) was reported in one patient (see Precautions).
Infections: In the clinical trial studying induction or remission (GPA/MPA study 1), which included 99 adult patients with severe GPA/MPA, the overall rate of infection was approximately 210 per 100 patient years (95% CI 173-256). Infections were predominately mild to moderate and consisted mostly of upper respiratory tract infections, herpes zoster and urinary tract infections. The rate of serious infections was approximately 25 per 100 patient years. The most frequently reported serious infection in the rituximab group was pneumonia at a frequency of 4%.
In the maintenance therapy clinical trial (GPA/MPA study 2) in adult patients, 30/57 (53%) patients in the rituximab arm and 33/58 (57%) in the azathioprine arm reported infections. The incidence of all grade infections was similar between the arms. Infections were predominately mild to moderate. The most common infections in the rituximab arm included upper respiratory tract infections, gastroenteritis, urinary tract infections and herpes zoster. The incidence of serious infections was similar in both arms (12%). The most commonly reported serious infection in the rituximab arm was mild or moderate bronchitis.
In the clinical studying pediatric GPA/MPA, the majority of reported infections were non-serious and predominately mild to moderate. The most common infections in the overall study period were: upper respiratory tract infections (URTIs) (48%), influenza (24%), conjunctivitis (20%), nasopharyngitis (20%), lower respiratory tract infections (16%), sinusitis (16%), viral URTIs (16%), ear infection (12%), gastroenteritis (12%) pharyngitis (12%), urinary tract infection (12%). Serious infections were reported in 7 patients (28%), and included: influenza (2 patients [8%]) and lower respiratory tract infection (2 patients [8%]) as the most frequently reported events.
Malignancies: In the clinical trial studying induction of remission (GPA/MPA study 1) the incidence of malignancy in rituximab treated patients was 2.05 per 100 patient years. On the basis of standardized incidence ratios, this malignancy rate appears to be similar to rates previously reported in GPA and MPA populations.
In the pediatric clinical trial, no malignancies were reported with a follow-up period of up to 54 months.
Laboratory Abnormalities: Hypogammaglobulinaemia (lgG or lgM below the lower limit of normal) has been observed in pediatric GPA/MPA patients treated with rituximab. During the overall study period, 3/25 (12%) patients reported an event of hypogammaglobulinaemia, 18 patients (72%) had prolonged low lgG levels (of whom 15 patients also had prolonged low lgM). Three patients received treatment with intravenous Immunoglobulin (IV-IG). There was no association between prolonged low lgG and lgM and an increased risk of serious infection.
Clinical Trial Experience in Pemphigus Vulgaris: Summary of the safety profile: The safety profile of rituximab in combination with short term, low dose, glucocorticoids in the treatment of patients with pemphigus vulgaris was studied in a randomized, controlled, multicenter, open-label study (PV study 1) in 38 pemphigus vulgaris (PV) and 8 pemphigus foliaceus (PF) patients. Patients randomized to the rituximab group received an initial 1000 mg IV on Study Day 1 and a second 1000 mg IV on Study Day 15. Maintenance doses of 500 mg IV were administered at Months 12 and 18. Patients could receive 1000 mg IV at the time of relapse (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
In the maintenance therapy clinical trial (PV study 2), a randomized, double-blind, double-dummy, active-comparator, multicenter study evaluating the efficacy and safety of rituximab compared with mycophenolate mofetil (MMF) in patients with moderate-to-severe PV requiring oral corticosteroids, 67 PV patients received treatment with rituximab (initial 1000 mg intravenous on Study Day 1 and a second 1000 mg intravenous on Study Day 15 repeated at Weeks 24 and 26) for up to 52 weeks (see PHARMACOLOGY: Pharmacodynamics: Clinical/Efficacy Studies under Actions).
The safety profile of rituximab in PV was consistent with the established safety profile in other approved autoimmune indications.
Tabulated list of adverse reactions for PV Studies 1 and 2: Adverse reactions from PV Studies 1 and 2 are presented in Table 29. In PV Study 1, ADRs were defined as adverse events which occurred at a rate of ≥ 5% among rituximab-treated PV patients, with a ≥ 2% absolute difference in incidence between the rituximab-treated group and the standard-dose prednisone group up to month 24. No patients were withdrawn due to ADRs in Study 1. In PV Study 2, ADRs were defined as adverse events occurring in ≥5% of patients in the rituximab arm and assessed as related. (See Table 29.)

Click on icon to see table/diagram/image

Further information on selected adverse drug reactions: Infusion-related reactions: In PV Study 1, infusion-related reactions were common (58%). Nearly all infusion-related reactions were mild to moderate. The proportion of patients experiencing an infusion-related reaction was 29% (11 patients), 40% (15 patients), 13% (5 patients), and 10% (4 patients) following the first, second, third, and fourth infusions, respectively. No patients were withdrawn from treatment due to infusion-related reactions. Symptoms of infusion related reactions were similar in type and severity to those seen in RA and GPA/MPA patients. In PV Study 2, IRRs occurred primarily at the first infusion and the frequency of IRRs decreased with subsequent infusions: 17.9%, 4.5%, 3% and 3% of patients experienced IRRs at the first, second, third, and fourth infusions, respectively. In 11/15 patients who experienced at least one IRR, the IRRs were Grade 1 or 2. In 4/15 patients, Grade ≥3 IRRs were reported and led to discontinuation of rituximab treatment; three of the four patients experienced serious (life-threatening) IRRs. Serious IRRs occurred at the first (2 patients) or second (1 patient) infusion and resolved with symptomatic treatment.
Infections: In PV Study 1, 14 patients (37%) in the rituximab group experienced treatment-related infections compared to 15 patients (42%) in the standard dose prednisone group. The most common infections in the rituximab group were herpes simplex and zoster infection, bronchitis, urinary tract infection, fungal infection, and conjunctivitis. Three patients (8%) in the rituximab group experienced a total of 5 serious infections (Pneumocystis jirovecii pneumonia, infective thrombosis, intervertebral discitis, lung infection, Staphylococcal sepsis) and one patient (3%) in the standard dose prednisone group experienced a serious infection (Pneumocystis jirovecii pneumonia).
In PV Study 2, 42 patients (62.7%) in the rituximab arm experienced infections. The most common infections in the rituximab group were upper respiratory tract infection, nasopharyngitis, oral candidiasis and urinary tract infection. Six patients (9%) in the rituximab arm experienced serious infections.
Laboratory Abnormalities: PV Study 2, in the rituximab arm, transient decreases in lymphocyte count, driven by decreases in the peripheral T-cell populations, as well as a transient decrease in phosphorus level were very commonly observed post-infusion. These were considered to be induced by intravenous methylprednisolone premedication infusion.
In PV Study 2, low IgG levels were commonly observed and low IgM levels were very commonly observed; however, there was no evidence of an increased risk of serious infections after the development of low IgG or IgM.
Hypogammaglobulinaemia (IgG and IgM below the lower limit of normal) has been observed in RA and adult and pediatric GPA/MPA patients treated with rituximab. There was no increased rate in overall infections or serious infections after the development of low IgG or IgM.
Rheumatoid Arthritis Patients: Events of neutropenia associated with rituximab treatment, the majority of which were transient and mild or moderate in severity, were observed in clinical trials in RA patients after the first course of treatment. Neutropenia can occur several months after the administration of rituximab.
In placebo-controlled periods of clinical trials, 0.94% (13/1382) of rituximab treated patients and 0.27% (2/731) of placebo patients developed severe (Grade 3 or 4) neutropenia. In these studies, rates of severe neutropenia were 1.06 and 0.53 per 100 patient years, respectively, after the first treatment course and 0.97 and 0.88 per 100 patient years, respectively, after multiple courses. Therefore, neutropenia can be considered an ADR for the first course only. Time to onset of neutropenia was variable. In clinical trials neutropenia was not associated with an observed increase in serious infection, and most patients continued to receive additional courses of rituximab after episodes of neutropenia.
Adult Patients with Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA) Patients: In the induction of remission clinical trial (GPA/MPA Study 1), at 6 months, in the rituximab group, 27%, 58% and 51% of patients with normal immunoglobulin levels at baseline, had low lgA, lgG and lgM levels, respectively compared to 25%, 50% and 46%, respectively in the cyclophosphamide group.
In the maintenance therapy clinical trial, no clinically meaningful differences between the two treatment arms or decreases in total immunoglobulin, lgG, lgM or lgA levels were observed throughout the trial.
In the induction of remission clinical trial, 24% of patients in the rituximab group (single course) and 23% of patients in the cyclophosphamide group developed CTC Grade 3 or greater neutropenia. Neutropenia was not associated with an observed increase in serious infection in rituximab treated patients.
Post Marketing: Non-Hodgkin's Lymphoma and Chronic Lymphocytic Leukaemia Patients: The reporting frequencies in this section (rare, very rare) are based on estimated marketed exposures and largely data derived from spontaneous reports. Additional cases of severe infusion-related reactions have been reported during post-marketing use of rituximab (see Precautions).
As part of the continuing post-marketing surveillance of rituximab safety, the following serious adverse reactions have been observed: Cardiovascular system: Severe cardiac events, including heart failure and myocardial infarction have been observed, mainly in patients with prior cardiac condition and/or cardiotoxic chemotherapy and mostly associated with infusion-related reactions. Vasculitis, predominantly cutaneous, such as leukocytoclastic vasculitis, has been reported very rarely.
Respiratory system: Respiratory failure/insufficiency and pulmonary infiltrates in the context of infusion-related reactions (see Precautions). In addition to pulmonary events associated with infusions, interstitial lung disease, some with fatal outcome, has been reported.
Blood and lymphatic system: Cases of infusion-related acute reversible thrombocytopenia have been reported.
Skin and appendages: Severe bullous skin reactions including fatal cases of toxic epidermal necrolysis have been reported rarely.
Nervous system: Cases of posterior reversible encephalopathy syndrome (PRES)/reversible posterior leukoencephalopathy syndrome (RPLS) have been reported. Signs and symptoms include visual disturbance, headache, seizures and altered mental status, with or without associated hypertension. A diagnosis of PRES/RPLS requires confirmation by brain imaging. The reported cases had recognized risk factors for PRES/RPLS, including the patients underlying disease, hypertension, immunosuppressive therapy and/or chemotherapy.
Cases of cranial neuropathy with or without peripheral neuropathy have been reported rarely. Signs and symptoms of cranial neuropathy, such as severe vision loss, hearing loss, loss of other senses and facial nerve palsy, occurred at various times up to several months after completion of rituximab therapy.
Body as a whole: Serum sickness-like reactions have been reported rarely.
Infections and infestations: Cases of hepatitis B reactivation have been reported, the majority of which were in subjects receiving rituximab in combination with cytotoxic chemotherapy (see Precautions). Other serious viral infections, either new, reactivation or exacerbation, some of which were fatal, have been reported with rituximab treatment. The majority of patients had received rituximab in combination with chemotherapy or as part of a haematopoietic stem cell transplant. Examples of these serious viral infections are infections caused by the herpes viruses (cytomegalovirus (CMV), Varicella zoster virus and Herpes simplex virus), JC virus (progressive multifocal leukoencephalopathy (PML), see Precautions) and Hepatitis C virus.
Progression of Kaposi's sarcoma has been observed in rituximab-exposed patients with pre-existing Kaposi's sarcoma. These cases occurred in non-approved indications and the majority of patients were HIV positive.
Gastro-intestinal system: Gastro-intestinal perforation, in some cases leading to death, has been observed in patients receiving rituximab in combination with chemotherapy for non-Hodgkin's lymphoma.
Rheumatoid Arthritis (RA), Granulomatosis with Polyangiitis (Wegener's) (GPA) and Microscopic Polyangiitis (MPA) Patients: As part of continuing post-marketing surveillance of rituximab safety, the following have been observed in the RA setting and are also expected, if not already observed, in GPA/MPA patients: Infections and infestations: Progressive multifocal leukoencephalopathy (PML) and reactivation of hepatitis B infection have been reported.
Body as a whole: Serum sickness-like reaction has been reported.
Skin and subcutaneous tissue disorders: Toxic epidermal necrolysis and Stevens-Johnson syndrome some with fatal outcome have been reported very rarely.
Blood and lymphatic system disorders: Neutropenic events, including severe late onset and persistent neutropenia have been reported rarely, some of which were associated with fatal infections.
Nervous system: Cases of posterior reversible encephalopathy syndrome (PRES)/reversible posterior leukoencephalopathy syndrome (RPLS) have been reported. Signs and symptoms include visual disturbance, headache, seizures and altered mental status, with or without associated hypertension. A diagnosis of PRES/RPLS requires confirmation by brain imaging. The reported cases had recognized risk factors for PRES/RPLS, including hypertension, immunosuppressive therapy and/or other concomitant therapies.
General disorders and administration site conditions: Severe IRRs some with fatal outcome have been reported (see Clinical Trials as previously mentioned).
Laboratory Abnormalities: Non-Hodgkin's Lymphoma: Blood and lymphatic system: Rarely the onset of neutropenia has occurred more than four weeks after the last infusion of rituximab. In post-marketing: studies of rituximab in patients with Waldenstrom's macroglobulinemia, transient increases in serum IgM levels have been observed following treatment initiation, which may be associated with hyperviscosity and related symptoms. The transient IgM increase usually returned to at least baseline level within 4 months.
Drug Interactions
At present, there are limited data on possible drug interactions with rituximab.
In CLL patients, co-administration with rituximab did not appear to have an effect on the pharmacokinetics of fludarabine or cyclophosphamide, in addition; there was no apparent effect of fludarabine and cyclophosphamide on the pharmacokinetics of rituximab.
Co-administration with methotrexate had no effect on the pharmacokinetics of rituximab in rheumatoid arthritis patients.
Patients with human anti-mouse antibody (HAMA) or human anti-chimeric antibody (HACA) titers may develop allergic or hypersensitivity reactions when treated with other diagnostic or therapeutic monoclonal antibodies.
In the RA clinical trial program, 373 rituximab treated patients received subsequent therapy with other disease-modifying antirheumatic drugs (DMARDs), of whom 240 received a biologic DMARD. In these patients the rate of serious infection while on rituximab (prior to receiving a biologic DMARD) was 6.1 per 100 patient years compared to 4.97 per 100 patient years following subsequent treatment with the biologic DMARD.
Caution For Usage
Special Instructions for Use, Handling and Disposal: Withdraw the required amount of Truxima under aseptic conditions and dilute to a calculated rituximab concentration of 1 - 4 mg/mL in an infusion bag containing sterile, non-pyrogenic 0.9%, aqueous saline solution or 5% aqueous dextrose solution. To mix the solution, gently invert the bag to avoid foaming. Care must be taken to ensure the sterility of prepared solution. Since the medicinal product does not contain any anti-microbial preservative or bacteriostatic agents, aseptic technique must be observed. Parenteral medications should be inspected visually for particulate matter or discoloration prior to administration.
The prepared infusion solution of Truxima is physically and chemically stable for 24 hours at 2°C - 8°C and subsequently 12 hours at room temperature.
Incompatibilities: No incompatibilities between rituximab and polyvinyl chloride or polyethylene bags or infusion sets have been observed.
Disposal of unused/expired medicines: The release of pharmaceuticals in the environment should be minimized. Medicines should not be disposed of via wastewater and disposal through household waste should be avoided. Use established "collection systems", if available in the location.
Storage
Store vials at 2°C - 8°C (in a refrigerator). Keep the container in the outer carton in order to protect from light.
After aseptic dilution in 0.9% aqueous saline solution: [Adding information following in-use stability study] The prepared infusion solution of rituximab in 0.9% aqueous saline solution is physically and chemically stable for 30 days at 2°C - 8°C and plus an additional 24 hours at room temperature.
After aseptic dilution in 5% aqueous dextrose solution: The prepared infusion solution of rituximab in 5% aqueous dextrose solution is physically and chemically stable for 24 hours at 2°C - 8°C plus an additional 12 hours at room temperature.
From a microbiological point of view, the prepared infusion solution should be used immediately. If not used immediately, in-use storage times and conditions prior to use are the responsibility of the user and would normally not be longer than 24 hours at 2°C - 8°C, unless dilution has taken place in controlled and validated aseptic conditions.
MIMS Class
Targeted Cancer Therapy
ATC Classification
L01FA01 - rituximab ; Belongs to the class of CD20 (Clusters of Differentiation 20) inhibitors. Used in the treatment of cancer.
Presentation/Packing
Form
Truxima conc for soln for infusion 10 mg/mL
Packing/Price
10 mL x 2 × 1's;50 mL x 1's
/thailand/image/info/truxima conc for soln for infusion 10 mg-ml/100 mg-10 ml?id=2a0bb425-c9bc-4e33-916b-ac07010aaab7
/thailand/image/info/truxima conc for soln for infusion 10 mg-ml/500 mg-50 ml?id=63f37d04-4101-4b08-9673-ac07010aaab7
Register or sign in to continue
Asia's one-stop resource for medical news, clinical reference and education
Already a member? Sign in
Register or sign in to continue
Asia's one-stop resource for medical news, clinical reference and education
Already a member? Sign in