Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Oct 1.
Published in final edited form as: Cancer. 2016 Jun 28;122(19):2996–3004. doi: 10.1002/cncr.30137

A Randomized Phase III Study in Low Grade Lymphoma Comparing Maintenance Anti-CD20 Antibody versus Observation following Induction Therapy: A trial of the ECOG-ACRIN Cancer Research Group (E1496)

Stefan K Barta 1, Hailun Li 2, Howard S Hochster 3, Fangxin Hong 2, Edie Weller 2, Randy D Gascoyne 4, Thomas M Habermann 5, Leo I Gordon 6, Natalia Colocci 7, Elizabeth M Bengtson 8, Sandra J Horning 9, Brad S Kahl 10
PMCID: PMC5030179  NIHMSID: NIHMS808030  PMID: 27351685

Abstract

Background

In the ECOG-ACRIN Cancer Research Group study, E1496, we reported that maintenance rituximab prolonged progression-free survival when compared to observation alone in patients with indolent lymphoma after induction chemotherapy. Here we present the long-term follow-up of the same patient cohort.

Methods

Patients with indolent lymphoma received induction chemotherapy with cyclophosphamide, vincristine, and prednisone (CVP). Patients with stable disease or better were then randomized to weekly rituximab (375mg/m2) ×4 every 6 months for 2 years (MR) or observation (OBS). The primary endpoint was progression-free survival (PFS); secondary endpoints were overall survival (OS), response rate and toxicities.

Results

Of 387 patients who initially received CVP induction, 158 were randomized to MR and 153 to OBS. After a median follow-up of 11.5 years, patients on MR had a longer median PFS (4.8 years) than patients on OBS (1.3 years; hazard ratio (HR) 0.49; p<0.0001). However, there was no difference in OS between MR and OBS (10-year OS 67% vs. 59%; median OS: 13.5 years vs. not reached; HR 0.91; p=0.69). Only minimal residual disease after induction therapy was significantly associated with PFS on multivariate analysis (HR 0.71; p=0.02). Low initial tumor burden, minimal residual disease, follicular histology, high FLIPI score, and male gender, were associated with OS. There was no difference in the rate of second primary malignancies between MR and OBS.

Conclusions

With long term follow up, MR did not influence the OS. The PFS benefit was maintained. MR should be considered optional in patients with indolent B-cell lymphoma.

Keywords: rituximab, rituximab maintenance, indolent lymphoma, follicular lymphoma, Non-Hodgkin lymphoma

Introduction

The CD20-directed monoclonal antibody rituximab was initially approved by the US FDA as a single agent for patients with indolent B-cell and follicular non-Hodgkin lymphoma (NHL) in 1997. When administered as a single agent on a weekly schedule of 375mg/m2 × 4 doses, the activity was relatively modest (progression-free survival [PFS] of 9.0 months). The complete response (CR) rate was 6% and the partial response (PR) rate was 42%, but the toxicity profile was very favorable (≤4% of ≥3 grade adverse events).1 Since then, studies have demonstrated that rituximab administered concurrently with a backbone of conventional chemotherapy, immunochemotherapy, not only leads to higher response rates and improved PFS compared to chemotherapy alone in the treatment of patients with follicular lymphoma both in the upfront and relapsed setting, but also prolongs overall survival (OS).24

Retrospective analyses of large cohort studies provide further evidence for the significant impact of rituximab on outcomes in indolent NHL. The median survival for patients with follicular lymphoma (FL), the most common indolent NHL, has improved over the last decade from 5–7 years to currently well over 10 years.59 This led to a paradigm shift in the approach to the treatment of indolent lymphomas, which are invariably characterized by their relapsing course. The previous dogma that choice of initial therapy had no impact on overall survival (OS) was revised. However, while the significant impact of rituximab in general is undisputed with combined immunochemotherapy the standard of care, the role of rituximab maintenance therapy after induction is less well defined.10

E1496 was one of the first randomized phase III studies to explore the role of rituximab as a maintenance strategy following induction chemotherapy in indolent lymphomas, albeit after induction with a chemotherapy only regimen. When first reported, median follow up was 3.7 years. Patients assigned to rituximab maintenance (MR) following cyclophosphamide, vincristine, and prednisone (CVP) induction therapy, had a 60% reduction in the risk of progression or death compared to observation (OBS) (p=0.44×10−10), and there was a trend for improved overall survival (relative risk [RR] of death 0.4; one-sided log-rank test p=0.05). Here we present the long-term results of E1496 with a median follow up of 11.5 years.

Material and Methods

Study Population

Patients aged 18 years or older with stage III or IV (Ann Arbor classification) small lymphocytic, follicular-small cleaved, or follicular mixed small cleaved and large cell lymphoma histology as per the Working Formulation (WF) classification were eligible for E1496. Patients with lymphoma that had both diffuse and follicular architectural elements were eligible as long as ≥50% of the cross-sectional areas were follicular. Additionally, as the WF lymphoma classification was revised during the study period, patients with marginal zone and lymphoplasmacytoid lymphoma became eligible. All cases underwent central pathology review by a hematopathologist (RDG). Patients were required to have measurable disease (lesions ≥2cm or splenomegaly on imaging), an ECOG performance status <2, and not have received prior lymphoma-directed (including radiation) therapy. The National Cancer Institute, ECOG, and the local IRB of each participating institution approved the protocol; informed consent was obtained according to institutional guidelines.

Study Design and Treatment

Study design and treatment were previously described in detail elsewhere.11 Briefly, patients were initially randomized to receive either intravenous (IV) cyclophosphamide 1000mg/m2 and vincristine 1.4mg/m2 (capped at 2.0mg) on day 1 as well as oral prednisone (100mg/m2) on days 1–5 of a 3-week cycle (CVP) to maximum response for at least 6 and at most 8 cycles; or IV cyclophosphamide 1000mg/m2 on day 1 and fludarabine 20mg/m2 on days 1–5 every 4 weeks (CF) for 4–6 cycles to best response. For this analysis, only patients enrolled on the CVP arm will be reported as the CF arm was closed early after an increased number of induction deaths had been observed (8 out of 115). Patients underwent CT imaging 4 weeks after completion of CVP or CF therapy to assess response. Patients with progressive disease (PD) as defined by a ≥25% increase in the sum of the products of the diameters of the pre-treatment lesions or new disease were taken off study. All other patients were randomized to either observation (OBS) or to receive maintenance rituximab (MR) with weekly IV rituximab (375mg/m2) × 4 every 6 months for a total of 2 years starting 4 weeks after completion of CVP or CF therapy. Randomization was stratified based on initial tumor burden (low tumor burden vs. high tumor burden defined as either ≥3 lymph nodes >3cm; one lesion >7cm; “B” symptoms; spleen size >16cm by CT scan; leukemic phase [>5000/mm3 atypical lymphocytes]; cytopenias; or risk of extrinsic compression of a vital organ), extent of residual disease after induction therapy (minimal residual disease vs. gross residual disease defined as either ≥10% bone marrow involvement or any lesion ≥2cm or with <75% size reduction), and histological subtype (follicular vs. other). CT imaging was repeated every 6 months for 2 years, then annually for 5 years; bone marrow biopsies were repeated annually for 5 years. This analysis is restricted to patients who were randomized to either MR or OBS after CVP induction.

Statistical Analysis

The primary end-point for the maintenance portion of the study was progression free survival (PFS) defined as time from maintenance randomization to death or lymphoma progression. OS was measured from random assignment to death as a result of any cause. After modifications following the early closure of the CF arm, the study required 172 failures and accrual estimate of 300 patients randomized to either MR or OBS for 83% power and a 5% one-sided type I error to detect a 50% increase in median PFS (2.5 years with OBS versus 3.75 years with MR). After 50% of events occurred, the study passed the O’Brien-Fleming boundary for positive results. Although accrual had been completed and all enrolled patients had been randomized by that time, investigators were given the option to treat OBS patients with MR. Comparisons between treatment groups were conducted according to the intent-to-treat principle among eligible patients with a stratified log-rank test. Three stratification factors were employed, including histology (follicular vs. other), residual disease after CVP (minimal or gross), and initial tumor burden (low or high). The Kaplan-Meier method and Cox proportional regression model were used to estimate failure rates, hazard ratios (HRs), and 95% confidence intervals (CIs). Toxicity was evaluated in all patients regardless of eligibility. Time-varying covariate was used in Cox proportional regression to test the proportional hazard assumption in OS evaluation.

Potential prognostic factors initial tumor burden, histology, residual disease status post CVP, FLIPI score, and gender were evaluated for associations with PFS and OS by univariate analyses using a log-rank test. Variables with a p-value ≤0.20 were then included in the multivariate model together with treatment arm with Cox proportional hazard model.

Results

Patient characteristics

Three hundred eighty seven patients were enrolled into the CVP induction. The overall response rate was 73% (60% PR, 13% CR). Following response assessment after CVP, 311 patients were randomized to the maintenance phase (Figure 1). Table 1 outlines the patient characteristics and demonstrates that patients on MR and OBS did not differ significantly in prognostic features with the exception of a higher prevalence of B-symptoms at baseline in patients assigned to MR (17% vs. 31%; p=0.005). Most patients had follicular histology (73%), high initial tumor burden (65%), high FLIPI scores (40%) and entered the maintenance phase with minimal residual disease (56%).

Figure 1.

Figure 1

Consort diagram for E1496

Table 1.

Characteristics of evaluable patients who entered the maintenance phase

All patients (n=311) Follicular lymphoma (n=228)
Characteristics All (%) MR n (%) OBS n (%) MR n (%) OBS n (%)
Age (median; range) 57 (26–86) 58 (26–84) 56 (32–86) 58 (30–84) 54 (32–80)
Male sex 171 (55) 86 (54) 85 (56) 59 (51) 62 (55)
ECOG PS 0 204 (66) 112 (71) 92 (60) 80 (70) 72 (64)
Histology: Follicular 228 (73) 115 (37) 113 (36) 115 (37) 113 (36)
Histology: non Follicular 83 (27) 43 (14) 40 (13)
 SLL/CLL 47 (15) 29 (9) 18 (6)
 LPL 2 (<1) 1 (<1) 1 (<1)
 Unclassifiable 7 (2) 3 (1) 4 (1)
 Other 15 (5) 6 (2) 9 (3)
 Missing 12 (4) 4 (1) 8 (3)
FLIPI score
 Low 57 (24) 28 (24) 29 (225) 23 (26) 24 (27)
 Intermediate 82 (35) 43 (36) 39 (34) 32 (36) 32 (36)
 High 95 (41) 48 (40) 47 (41) 33 (38) 33 (37)
Stages IV 217 (70) 109 (69) 108 (71) 73 (64) 72 (64)
Elevated LDHa 53 (23) 29 (25) 24 (21) 24 (27) 19 (22)
High tumor burden 200 (64) 96 (61) 104 (68) 69 (60) 75 (66)
Minimal residual disease 175 (56) 89 (56) 86 (56) 67 (58) 65 (58)
Response to CVP
 Complete response 49 (16) 25 (16) 24 (16) 18 (16) 15 (13)
 Partial response 210 (68) 110 (69) 100 (65) 81 (70) 77 (68)
 Stable disease 40 (13) 17 (11) 23 (15) 12 (10) 17 (15)
 Not evaluable 12 (4) 6 (4) 6 (4) 4 (4) 4 (4)
Response on maintenanceb
 Complete response 92 (30) 59 (37) 33 (22) 43 (37) 24 (21)
 Partial response 170 (55) 78 (49) 92 (60) 60 (52) 68 (60)
 Stable disease 37 (12) 15 (9) 22 (14) 8 (7) 17 (15)
 Not evaluable 12 (4) 6 (4) 6 (4) 4 (3) 4 (4)

Abbreviations: MR, maintenance rituximab; OBS, observation; SLL/CLL, small lymphocytic lymphoma/chronic lymphocytic leukemia; LPL, lymphoplasmacytic lymphoma.

a

Not available for all patients.

b

Response on maintenance was defined as best response during the maintenance phase

During the 2-year maintenance phase, 22% (n=35) of MR patients improved their response from PR to CR (n=33) or from SD to PR or CR (n=1 each). In comparison, significantly fewer OBS patients (7%), (n=10; p=0.00006) demonstrated improvement in their response (PR to CR, (n=9); SD to PR, (n=1)). As previously reported, grade 3 and 4 toxcicites during the maintenance phase were generally rare and not significantly different between MR and OBS except for grade 4 neutropenia (2% vs. 0%).11

Maintenance: survival results

After a median follow-up of 11.5 years, the median PFS for the MR arm among all evaluable patients was 4.8 years compared to 1.3 years for patients on the OBS arm (p<0.0001). The stratified hazard ratio (HR) was 0.49 (95% confidence interval (CI), 0.37–0.64). The results amongst patients with FL histology were similar, with a median PFS of 4.6 and 1.3 years respectively for the MR and OBS arm (stratified HR 0.46; 95% CI, 0.34–0.63; p<0.0001). Figure 2 shows the PFS for all evaluable patients and patients with FL histology. The improvement in PFS was maintained throughout the entire follow-up period, with 5 and 10 year PFS rates of 47% and 26% for MR vs. 26% and 9% for OBS, respectively (Table 2). The results were similar in the subset of patients with follicular lymphoma (supplemental Table 1S).

Figure 2.

Figure 2

Progression-free survival for a) all evaluable patients (n=311) and b) patients with follicular lymphoma (n=228)

Table 2.

Survival probabilities all evaluable patients (n=311)

MR (n=158) OBS (n=153)
PFS (95% CI)
 3 years 0.66 (0.58–0.73) 0.35 (0.28–0.43)
 5 years 0.47 (0.39–0.55) 0.26 (0.19–0.34)
 10 years 0.26 (0.17–0.36) 0.09 (0.04–0.15)
 Median PFS 4.8 years 1.3 years
OS (95% CI)
 3 years 0.92 (0.87–0.96) 0.86 (0.80–0.91)
 5 years 0.85 (0.78–0.89) 0.78 (0.71–0.84)
 10 years 0.67 (0.58–0.73) 0.59 (0.51–0.67)
 Median OS 13.5 years Not reached

Abbreviations: MR, maintenance rituximab; OBS, observation; PFS, progression-free survival; 95% CI, 95% confidence interval, OS, overall survival.

The OS curves crossed at approximately 12 years, however the interaction between the treatment groups and time was not statistically significant (p=0.1) which suggests that the proportional hazard assumption was not violated. Therefore, no statistical significant difference in OS was observed by stratified log-rank test (p=0.41). The median OS was 13.5 years for the MR arm, and has not been reached for the OBS arm. The stratified hazard ratio (HR) was 0.86 (95% CI, 0.61–1.23). Extended Cox regression with time-varying covariates revealed a stratified HR for MR over OBS of 0.80 (95%CI, 0.56–1.15) before 12 years and 5.61 (95% CI, 0.58–46.3) after 12 years. Neither time interval difference was significant (p=0.23 and p=0.16, respectively), which further confirms that there is no significantly difference in OS between MR and OBS The results among patients with FL histology were similar, with a median OS of 13.5 years for patients receiving MR and not reached for patients on the OBS arm (stratified HR=0.91; 95% CI, 0.59–1.42; p=0.69). The interaction between the treatment groups and time was statistically significant (p=0.05) in the Cox model. However, extended Cox regression with time-varying covariates revealed a stratified HR for MR over OBS of 0.81 (95%CI, 0.51–1.28) before 12 years and 6.65 (95% CI, 0.70–63.0) after 12 years. Neither time interval difference was significant (p=0.38 and p=0.10, respectively). Figure 2a and 2b demonstrate the OS for all evaluable patients and the FL subset, respectively. The Kaplan-Meier estimates of survival rate at different time points are shown in Table 2 (Supplemental Table 2S for the follicular subset).

Patients experienced an approximately 50% improvement in PFS with MR versus OBS irrespective of initial tumor burden, residual disease after CVP induction or lymphoma histology (Table 3). There was no difference in OS between the subgroups. (Kaplan-Meier curves for the subgroups can be found in the supplemental material). When the analysis was restricted to only female patients, there was no difference in OS between MR and OBS. There was no significant interaction between the variables maintenance assignment and gender.

Table 3.

Progression-free and overall survival for MR vs. OBS by subgroups

PFS OS
10-yr PFS
MR vs OBS
Hazard Ratio
(95% CI)
P 10-yr OS
MR vs OBS
Hazard Ratio
(95% CI)
P
All Patients (n=311) 0.26 vs 0.09 0.49 (0.37–0.64) <0.0001 0.67 vs 0.59 0.86 (0.61–1.23) 0.41
Tumor burden
Low (n=111) 0.38 vs 0.11 0.46 (0.29–0.74) 0.006 0.79 vs 0.74 1.01 (0.50–2.04) 0.98
High (n=200) 0.20 vs 0.08 0.50 (0.36–0.69) <0.0001 0.59 vs 0.52 0.82 (0.54–1.23) 0.33
Histology
Follicular (n=228) 0.28 vs 0.08 0.46 (0.34–0.63) <0.0001 0.72 vs 0.65 0.91 (0.59–1.42) 0.69
Other (n=83) 0.20 vs 0.12 0.57 (0.35–0.94) 0.03 0.53 vs 0.43 0.83 (0.47–1.47) 0.51
Residual disease
Minimal (n=175) 0.27 vs 0.12 0.51 (0.35–0.74) 0.0004 0.74 vs 0.66 0.81 (0.48–1.37) 0.43
Gross (n=136) 0.23 vs 0.05 0.47 (0.33–0.68) <0.0001 0.57 vs 0.50 0.91 (0.56–1.46) 0.68

Abbreviations: MR, maintenance rituximab; OBS, observation; PFS, progression-free survival; 95% CI, 95% confidence interval, OS, overall survival.

*

Hazard ratios were based on Cox proportional hazards models stratified by all the stratification factors used in randomization (tumor burden, histology, and residual disease) except the one being analyzed. P-value was based on stratified log rank-test.

Prognostic factors

Table 4 lists the results of the univariate and multivariate analyses of prognostic factors in all evaluable patients (n=311). Low initial tumor burden (HR 0.68; p=0.005), minimal residual disease (HR 0.35; p=0.0005) and complete response to CVP induction (HR 0.58; p=0.005) were associated on univariate analysis with improved PFS, but histology, FLIPI, or sex were not. On multivariate analysis, only minimal residual disease on imaging remained statistically significantly associated with PFS (HR 0.71; 95%CI 0.54–0.94; p=0.02), while low tumor burden and response to CVP showed a trend towards longer PFS (HR 0.79, p=0.11; and HR=0.67, p=0.06, respectively). MR significantly improved PFS compared to OBS (HR=0.47, p<0.0001) after adjusting for other prognostic factors in the multivariate model. For OS, low tumor burden (HR 0.52; p=0.0009), minimal residual disease (HR 0.51; p=0.0001), follicular histology (HR 0.56; p=0.001), response to CVP (HR 0.57, p=0.04) and female gender (HR 0.65, p=0.02) were associated on prolonged OS, while high FLIPI scores (HR 1.77; p=0.001) predicted for worse OS on univariate analysis. In the multivariate model, all factors except response to CVP remained significantly associated with OS, while treatment arm (MR vs. OBS) remained non-significant.

Table 4.

Univariate and multivariate analysis of prognostic factors in all evaluable patients (n=311)

PFS OS
Univariate Analysis Multivariate Analysis Univariate Analysis Multivariate Analysis
Hazard Ratio (95% CI) P Hazard Ratio (95% CI) P Hazard Ratio (95% CI) P Hazard Ratio (95% CI) P
Treatment Arm 0.49
(0.37–0.63)
<.0001 0.47
(0.36–0.61)
<.0001 0.90
(0.64–1.27)
0.56 0.88
(0.63–1.25)
0.49
MR (n=158) vs OBS (n=153)
Tumor burden 0.68
(0.51–0.89)
0.005 0.79
(0.59–1.06)
0.11 0.52
(0.35–0.77)
0.0009 0.64
(0.43–0.96)
0.03
Low (n=111) vs. High (n=200)
Histology 0.99
(0.74–1.31)
0.92 0.56
(0.39–0.80)
0.001 0.56
(0.39–0.81)
0.002
Follicular (n=228) vs. Other (n=83)
Residual disease 0.63
(0.49–0.82)
0.0005 0.71
(0.54–0.94)
0.02 0.51
(0.36–0.73)
0.0001 0.62
(0.42–0.90)
0.01
Minimal (n=175) vs. Gross (n=136)
Response to CVP 0.58
(0.39–0.85)
0.005 0.67
(0.44–1.02)
0.06 0.57
(0.33–0.98)
0.04 0.78
(0.44–1.41)
0.41
CR (n=49) vs. Other (n=262)
FLIPI 1.05
(0.79–1.38)
0.75 1.77
(1.24–2.52)
0.001 1.75
(1.21–2.53)
0.003
High risk (n=95) vs. Other (n=216)
Gender 0.85
(0.66–1.11)
0.23 0.65
(0.46–0.93)
0.02 0.58
(0.40–0.83)
0.003
Female (n=140) vs. Male (n=171)

Abbreviations: HR, hazard ratio; 95% CI, 95% confidence interval

Second primary malignancies

A total of 63 second primary cancers developed in 55 patients; 6 patients had multiple second malignancies (Supplemental Table 3S). Based on the Fisher’s exact test, significantly less patients developed secondary primary cancer in the MR arm compared to the OBS arm (p=0.03), with 25 cancers occurring in 20 patients on MR versus 38 cancers in 35 patients on OBS. The 2 most common second malignancy were lung cancer (n=15) and non-melanomatous skin cancer (n=10). Hematological malignancies occurred in 1 patient on the MR arm (Hodgkin lymphoma (HL)) and 5 patients on the OBS arm (n=1 each for FL, reticulosarcoma, HL, chronic myelogenous leukemia (CML), and myelodysplastic syndrome (MDS).

Discussion

The long-term follow-up results for E1496 after a median follow up of 11.5 years demonstrate a substantial improvement in PFS with rituximab maintenance (MR) after induction therapy with CVP in patients with indolent B-cell lymphomas. With more than one decade after the completion of induction chemotherapy, an approximately 50% reduction in the composite outcome of lymphoma PFS was observed in the MR group, although no improvement in OS.

The classical prognostic factors for OS, such as the FLIPI score, initial tumor burden, and response to induction treatment,1214 were significant in E1496 study. The PFS benefit of MR was similar in all subgroups, irrespective of initial tumor burden, response to induction chemotherapy, or histological subtype. There was no increase in secondary primary malignancies in patients treated with MR, and toxicities were generally manageable as previously described.11 After 50% of the events had occurred, investigators had the option to treat patients randomized to OBS with MR. Cross-over therefore could have attenuated the difference on PFS and OS. Unfortunately information on second-line treatment was not available, which furthermore might have confounded our findings.

Our findings are consistent with the published literature. Rituximab maintenance following upfront systemic therapy was first formally assessed by Hainsworth and colleagues in a multi-center phase 2 trial. Sixty patients with indolent NHL (follicular and small lymphocytic lymphoma) received induction with 4 weekly doses of rituximab. Patients with SD or better then proceeded to maintenance with 4 further courses of weekly rituximab x4 every 6 months, similar to E1496. The observed actuarial median PFS of 34 months constituted a significant improvement compared to historical data for rituximab induction alone.15 Ghielmini and colleagues on behalf of the Swiss Group for Clinical Cancer Research (SAKK) confirmed in SAKK35/98 that rituximab maintenance following rituximab induction with 4 weekly doses improved the PFS when administered as a single dose every 2 months ×4 versus observation alone in untreated patients with FL (19 vs. 36 months; p=0.009).16 An unspecified number of patients in these studies had low tumor burden disease when based on accepted criteria, such as the GELF criteria,14 and might not be considered candidates for induction chemotherapy. Our study also involved low and high tumor burden patients, and the PFS benefit of MR was seen in both groups. Not surprisingly, we also report a longer OS in patients with low tumor burden compared to high tumor burden patients.

In addition to E1496, the only other large randomized phase III trial assessing MR after chemotherapy-based induction is the PRIMA trial, which reported at a median follow-up of 36 months a 45% improvement in PFS (HR 0.55; 95%CI 0.44–0.68) without an OS benefit for patients on MR.17 In this study by the Groupe d’Etude des Lymphomes de l’Adulte (GELA), 1217 patients with previously untreated high-tumor burden FL received induction therapy with either R-CVP, R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) or R-FCM, following which patients with CR or PR were randomized to MR (12 infusions, one every 2 months; n=505) or observation (n=513). As in E1496, the PFS benefit was observed in all patient subgroups independent of FLIPI score, age, sex, or response to induction therapy. In the PRIMA study, only 28 patients received R-FCM for induction, and there were no conclusions about the benefit in this subgroup. Following R-CHOP induction, patients experienced a borderline increased PFS benefit compared to R-CVP induction (HR 0.51 (95%CI 0.39–0.65) vs. HR 0.69 (95%CI 0.41–1.02)). The PFS benefit of MR in E1496 is similar to the PRIMA study, where all patients also received rituximab in the induction treatment.

Considering the median survival for patients with indolent lymphomas, and specifically FL, which currently exceeds 10 years, long-term follow-up for interventional therapeutic studies in this setting is of utmost importance to determine whether improvement in surrogate markers for OS, such as response rates or PFS, eventually translate into an OS benefit.7,18 In the SAKK 35/98 trial at a median follow-up 9.5 years, there was a persistent event-free survival (EFS) benefit in untreated patients (HR 0.59; 95%CI 0.39–0.88), which is comparable to E1496. When comparing both studies it needs to be noted that in SAKK 35/98 the primary outcomes was EFS, defined as time from first induction to either progression, relapse, second tumor, or death from any cause, as opposed to PFS, defined in E1496 as time to death or lymphoma progression. While 45% of patient in the MR arm were still alive at 8 years without events, there was no OS benefit.19 However, given the low death rate, the significant impact of salvage therapy on survival, and the fact that patients might have received MR later, influencing OS might be an unachievable objective with a therapeutic intervention early in the disease course when the therapeutic intervention eventually will become available to all patients. This might be different in a later setting. A meta-analysis examining the benefit of MR found improved OS in the relapsed setting (HR for death 0.58; 95%CI 0.42–0.79), while no OS benefit was apparent for MR in the upfront setting.20

Considerations that have to be taken into account when evaluating the PFS gain with MR are safety, quality of life, and cost-effectiveness. In the PRIMA trial that incorporated initial immunochemotherapy, toxicities in patients on MR were mainly grade 2–4 infections (39% vs. 24%; HR 1.62; p<0.0001), although no differences between MR and OBS in immunoglobulin levels were seen. The most common infections affected the upper airways and urinary tract.17 A meta-analysis described an infection-related adverse event rate that was double in MR patients (HR 1.99; 1.21–3.27; p=0.007).20 Other immediate and late toxicities are cytopenias, specifically neutropenia (5–10%),1517,21 and reactivation of hepatitis B or C, which can occur late, but has not yet been systematically quantified.22 Most of these toxicities are considered mild to moderate and manageable. We and others did not find an increase in second cancers.19 The influence of MR on the rate of histological transformation remains unclear, as information on histological transformation was not routinely collected during follow up for E1496. While there is controversy about improvement in quality of life with rituximab maintenance,17,23,24 some studies suggest an economic benefit.2527

In conclusion, first-line MR for indolent lymphomas following chemotherapy with CVP results in a substantial and sustained PFS improvement with an acceptable safety profile irrespective of initial tumor burden or response to induction therapy. Progression free survival was nearly 3 times longer with maintenance rituximab (4.8 vs 1.3 years) and at 3 years nearly twice as many patients had not progressed (66% vs 35% progression free). Nonetheless, even after median follow up of 11.5 years, there is no benefit in overall survival as the curves come back together after 10 years. This suggests a prolonged effect on remission, but eventual salvage with subsequent therapy. Furthermore, scientific questions that remain unanswered are the optimal schedule and length of rituximab maintenance, the benefit of MR following induction therapy compared to a rituximab retreatment “as-needed” approach as pioneered by the RESORT trial.28 The long-term follow up data from E1496 demonstrate that MR has a long-lasting impact on PFS without long-term toxicity, making it an attractive option for patients. Given the lack of impact on OS, MR should be considered an optional intervention in the management of indolent lymphoma.

Supplementary Material

Suppl

Figure 3.

Figure 3

Overall survival for a) all evaluable patients (n=311) and b) patients with follicular histology (n=228)

Acknowledgments

Funding: This study was coordinated by the ECOG-ACRIN Cancer Research Group (Robert L. Comis, MD and Mitchell D. Schnall, MD, PhD, Group Co-Chairs) and supported in part by Public Health Service Grants CA180794, CA180820, CA180826, CA180790, CA180816, CA180799, CA180821 and from the National Cancer Institute, National Institutes of Health and the Department of Health and Human Services. Its content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute.

Footnotes

Conflict of interest disclosures: S.H. is an employee of Genentech; the authors report no other relevant conflicts of interests

References

  • 1.McLaughlin P, Grillo-Lopez AJ, Link BK, et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J Clin Oncol. 1998;16(8):2825–2833. doi: 10.1200/JCO.1998.16.8.2825. [DOI] [PubMed] [Google Scholar]
  • 2.Marcus R, Imrie K, Solal-Celigny P, et al. Phase III study of R-CVP compared with cyclophosphamide, vincristine, and prednisone alone in patients with previously untreated advanced follicular lymphoma. J Clin Oncol. 2008;26(28):4579–4586. doi: 10.1200/JCO.2007.13.5376. [DOI] [PubMed] [Google Scholar]
  • 3.Hiddemann W, Kneba M, Dreyling M, et al. Frontline therapy with rituximab added to the combination of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) significantly improves the outcome for patients with advanced-stage follicular lymphoma compared with therapy with CHOP alone: results of a prospective randomized study of the German Low-Grade Lymphoma Study Group. Blood. 2005;106(12):3725–3732. doi: 10.1182/blood-2005-01-0016. [DOI] [PubMed] [Google Scholar]
  • 4.Forstpointner R, Dreyling M, Repp R, et al. The addition of rituximab to a combination of fludarabine, cyclophosphamide, mitoxantrone (FCM) significantly increases the response rate and prolongs survival as compared with FCM alone in patients with relapsed and refractory follicular and mantle cell lymphomas: results of a prospective randomized study of the German Low-Grade Lymphoma Study Group. Blood. 2004;104(10):3064–3071. doi: 10.1182/blood-2004-04-1323. [DOI] [PubMed] [Google Scholar]
  • 5.Fisher RI, LeBlanc M, Press OW, Maloney DG, Unger JM, Miller TP. New treatment options have changed the survival of patients with follicular lymphoma. J Clin Oncol. 2005;23(33):8447–8452. doi: 10.1200/JCO.2005.03.1674. [DOI] [PubMed] [Google Scholar]
  • 6.Nabhan C, Aschebrook-Kilfoy B, Chiu BC, Kruczek K, Smith SM, Evens AM. The impact of race, age, and sex in follicular lymphoma: A comprehensive SEER analysis across consecutive treatment eras. Am J Hematol. 2014;89(6):633–638. doi: 10.1002/ajh.23708. [DOI] [PubMed] [Google Scholar]
  • 7.Tan D, Horning SJ, Hoppe RT, et al. Improvements in observed and relative survival in follicular grade 1–2 lymphoma during 4 decades: the Stanford University experience. Blood. 2013;122(6):981–987. doi: 10.1182/blood-2013-03-491514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Friedberg JW, Byrtek M, Link BK, et al. Effectiveness of first-line management strategies for stage I follicular lymphoma: analysis of the National LymphoCare Study. J Clin Oncol. 2012;30(27):3368–3375. doi: 10.1200/JCO.2011.40.6546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Swenson WT, Wooldridge JE, Lynch CF, Forman-Hoffman VL, Chrischilles E, Link BK. Improved survival of follicular lymphoma patients in the United States. J Clin Oncol. 2005;23(22):5019–5026. doi: 10.1200/JCO.2005.04.503. [DOI] [PubMed] [Google Scholar]
  • 10.Friedberg JW. Rituximab maintenance in follicular lymphoma: PRIMA. The Lancet. 377(9759):4–6. doi: 10.1016/S0140-6736(10)62272-6. [DOI] [PubMed] [Google Scholar]
  • 11.Hochster H, Weller E, Gascoyne RD, et al. Maintenance rituximab after cyclophosphamide, vincristine, and prednisone prolongs progression-free survival in advanced indolent lymphoma: results of the randomized phase III ECOG1496 Study. J Clin Oncol. 2009;27(10):1607–1614. doi: 10.1200/JCO.2008.17.1561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bachy E, Brice P, Delarue R, et al. Long-term follow-up of patients with newly diagnosed follicular lymphoma in the prerituximab era: effect of response quality on survival–A study from the groupe d’etude des lymphomes de l’adulte. J Clin Oncol. 2010;28(5):822–829. doi: 10.1200/JCO.2009.22.7819. [DOI] [PubMed] [Google Scholar]
  • 13.Solal-Celigny P, Roy P, Colombat P, et al. Follicular lymphoma international prognostic index. Blood. 2004;104(5):1258–1265. doi: 10.1182/blood-2003-12-4434. [DOI] [PubMed] [Google Scholar]
  • 14.Solal-Celigny P, Lepage E, Brousse N, et al. Doxorubicin-containing regimen with or without interferon alfa-2b for advanced follicular lymphomas: final analysis of survival and toxicity in the Groupe d’Etude des Lymphomes Folliculaires 86 Trial. J Clin Oncol. 1998;16(7):2332–2338. doi: 10.1200/JCO.1998.16.7.2332. [DOI] [PubMed] [Google Scholar]
  • 15.Hainsworth JD, Litchy S, Burris HA, 3rd, et al. Rituximab as first-line and maintenance therapy for patients with indolent non-hodgkin’s lymphoma. J Clin Oncol. 2002;20(20):4261–4267. doi: 10.1200/JCO.2002.08.674. [DOI] [PubMed] [Google Scholar]
  • 16.Ghielmini M, Schmitz S-FH, Cogliatti SB, et al. Prolonged treatment with rituximab in patients with follicular lymphoma significantly increases event-free survival and response duration compared with the standard weekly × 4 schedule. 1032004 doi: 10.1182/blood-2003-10-3411. [DOI] [PubMed] [Google Scholar]
  • 17.Salles G, Seymour JF, Offner F, et al. Rituximab maintenance for 2 years in patients with high tumour burden follicular lymphoma responding to rituximab plus chemotherapy (PRIMA): a phase 3, randomised controlled trial. Lancet. 2011;377(9759):42–51. doi: 10.1016/S0140-6736(10)62175-7. [DOI] [PubMed] [Google Scholar]
  • 18.Pulte D, Gondos A, Brenner H. Ongoing improvement in outcomes for patients diagnosed as having Non-Hodgkin lymphoma from the 1990s to the early 21st century. Arch Intern Med. 2008;168(5):469–476. doi: 10.1001/archinternmed.2007.125. [DOI] [PubMed] [Google Scholar]
  • 19.Martinelli G, Hsu Schmitz S-F, Utiger U, et al. Long-Term Follow-Up of Patients With Follicular Lymphoma Receiving Single-Agent Rituximab at Two Different Schedules in Trial SAKK 35/98. Journal of Clinical Oncology. 2010;28(29):4480–4484. doi: 10.1200/JCO.2010.28.4786. [DOI] [PubMed] [Google Scholar]
  • 20.Vidal L, Gafter-Gvili A, Leibovici L, et al. Rituximab maintenance for the treatment of patients with follicular lymphoma: systematic review and meta-analysis of randomized trials. J Natl Cancer Inst. 2009;101(4):248–255. doi: 10.1093/jnci/djn478. [DOI] [PubMed] [Google Scholar]
  • 21.van Oers MH, Klasa R, Marcus RE, et al. Rituximab maintenance improves clinical outcome of relapsed/resistant follicular non-Hodgkin lymphoma in patients both with and without rituximab during induction: results of a prospective randomized phase 3 intergroup trial. Blood. 2006;108(10):3295–3301. doi: 10.1182/blood-2006-05-021113. [DOI] [PubMed] [Google Scholar]
  • 22.Seto WK, Chan TS, Hwang YY, et al. Hepatitis B reactivation in patients with previous hepatitis B virus exposure undergoing rituximab-containing chemotherapy for lymphoma: a prospective study. J Clin Oncol. 2014;32(33):3736–3743. doi: 10.1200/JCO.2014.56.7081. [DOI] [PubMed] [Google Scholar]
  • 23.Ardeshna KM, Qian W, Smith P, et al. Rituximab versus a watch-and-wait approach in patients with advanced-stage, asymptomatic, non-bulky follicular lymphoma: an open-label randomised phase 3 trial. The Lancet. Oncology. 2014;15(4):424–435. doi: 10.1016/S1470-2045(14)70027-0. [DOI] [PubMed] [Google Scholar]
  • 24.Wagner LI, Zhao F, Hong F, et al. Anxiety and Health-Related Quality of Life Among Patients With Low-Tumor Burden Non-Hodgkin Lymphoma Randomly Assigned to Two Different Rituximab Dosing Regimens: Results From ECOG Trial E4402 (RESORT) J Clin Oncol. 2015;33(7):740–748. doi: 10.1200/JCO.2014.57.6801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Blommestein HM, Issa DE, Pompen M, et al. Cost-effectiveness of rituximab as maintenance treatment for relapsed follicular lymphoma: results of a population-based study. Eur J Haematol. 2014;92(5):398–406. doi: 10.1111/ejh.12264. [DOI] [PubMed] [Google Scholar]
  • 26.Deconinck E, Miadi-Fargier H, Pen CL, Brice P. Cost effectiveness of rituximab maintenance therapy in follicular lymphoma: long-term economic evaluation. PharmacoEconomics. 2010;28(1):35–46. doi: 10.2165/11314070-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 27.Greenhalgh J, Bagust A, Boland A, et al. Rituximab for the first-line maintenance treatment of follicular non-Hodgkin’s lymphoma : a NICE single technology appraisal. PharmacoEconomics. 2013;31(5):403–413. doi: 10.1007/s40273-013-0043-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kahl BS, Hong F, Williams ME, et al. Rituximab extended schedule or re-treatment trial for low-tumor burden follicular lymphoma: eastern cooperative oncology group protocol e4402. J Clin Oncol. 2014;32(28):3096–3102. doi: 10.1200/JCO.2014.56.5853. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Suppl

RESOURCES