Abstract

Treatment for follicular lymphoma (FL) has rapidly advanced in recent years, with a broad array of novel agents demonstrating promising results, particularly in the relapsed or refractory (R/R) setting. Key emerging therapies include chimeric antigen receptor T-cell therapy, bispecific antibodies, and various targeted therapies, many of which are currently being evaluated in frontline and earlier lines of relapses, against standard of care. These new therapeutic approaches offer the potential for improved patient outcomes, but their integration into clinical practice presents challenges such as determining the optimal sequencing and selection of treatment, balancing unique benefits, toxicities, and treatment burdens, and overall therapeutic goals, especially in the R/R space. Moreover, the heterogeneous nature of FL, coupled with its indolent and long-term course, necessitates a personalized approach to therapy that considers the individual patient’s needs and preferences. This approach ensures that the most suitable treatment is chosen, balancing efficacy with quality-of-life considerations. In this review, we provide a comprehensive summary and in-depth discussion of ongoing phase 3 clinical trials both at frontline and R/R. We explore various aspects of these trials, including their study designs, limitations, and potential clinical applicability. In addition, we examine how these trials may shape future treatment practices and address the broader challenges surrounding the concept of achieving a cure in FL. By critically analyzing the evolving landscape of FL treatment, this review aims to offer valuable insights into how these new therapeutic strategies might affect patient management, and the future direction of FL clinical studies.

Follicular lymphoma (FL) is the most prevalent indolent B-cell non-Hodgkin lymphoma (B-NHL) worldwide, representing ∼20% of all NHL cases. In the United States, ∼14 000 new cases are diagnosed annually, making it a relevant concern for patients and health care providers.1,2 Historically, FL has been considered incurable despite advances in treatment; however, some patients do achieve prolonged disease control. FL follows a relapsing-remitting course, with continuous patterns of recurrence and progressively shorter periods of disease control after each line of treatment.3 In addition, its high heterogenicity with varying clinical behavior makes management particularly nuanced.

Over the past 2 decades, the treatment landscape for FL has evolved significantly, particularly since the introduction of rituximab and other novel therapies. Rituximab marked a ground-breaking advance, but it is no longer a singular solution.4 Today, new therapeutic approaches are being explored, including novel immunotherapies, cellular therapies, and targeted therapies that aim to selectively eliminate cancer cells while minimizing damage to healthy tissues. There is growing discussion around the potential curability of FL, challenging the traditional view of the disease as incurable. Despite the broadening array of treatment options, determining the optimal sequence of therapies for FL remains a major challenge for clinicians, particularly for relapsed or refractory (R/R) FL.

In this review, we explore the current landscape of FL therapies, focusing on ongoing phase 3 trials that may provide clearer guidance on the most effective treatment strategies moving forward.

Limited-stage disease FL

For limited-stage FL, defined as stage I or contiguous stage II, 24 Gy of local radiotherapy (RT) is associated with durable remission and is considered the standard of care (SOC).5 The Follicular Radiotherapy Trial (FoRT), which compared 24 Gy to 4 Gy in the frontline setting among 548 patients, demonstrated that lower dose of 4 Gy resulted in inferior local progression-free survival (PFS), with an estimated 3-year PFS difference of 19.8%.6 This finding suggests that reducing the radiation dose compromises local disease control and spurred interest in combining systemic therapy with RT in this patient population.

Building on the FoRT study concept, the FORTplus (NCT05045664) trial aims to evaluate whether the use of a more potent CD20 monoclonal antibody (mAb) can allow for a reduction in total RT dose.7 This multicenter study randomizes patients with limited-stage disease to receive either 24 Gy RT plus rituximab or 4 Gy RT plus obinutuzumab. The primary end point is complete remission rate (CRR), with PFS and overall survival (OS) as secondary end points. Obinutuzumab, considered a more potent CD20 mAb, may potentially offset the effects of reduced radiation dosing Table 1.

Table 1.

Features of ongoing phase 3 frontline FL trials

NameTrialEnrollment targetPatientsPrimary end pointImportant secondary end points
Limited stage 
FORTplus (NCT05045664)7  RT 24 Gy + rituximab vs 4 Gy + obinutuzumab 100 Age, ≥18 years
Stage IA or IIA, nonbulky (≤7 cm)
Exclude extranodal 
CRR based on CT scan Metabolic CRR
PFS
OS
Rate of AEs 
FIL_GAZEBO (NCT05929222)8  RT 24 Gy vs 24 Gy + obintuzumab 190 Age, ≥18 years
Stage IA or IIA, or IE, nonbulky (<7 cm)
FLIPI of <2, FLIPI2 of ≤2 
PFS CRR
ORR
MRD negativity
Rate of AEs 
Low tumor burden 
SWOG2308 (NCT06337318)23  Mosunetuzumab vs rituximab 600 Age, ≥18 years
Define low tumor burden: not fulfill GELF criteria
Participants must either be experiencing distress due to disease or prefer active management rather than watch and wait
Allow patients with stage I disease who decline/are not suitable for RT 
PFS
3-year PFS 
OS
ORR
Rate of AEs 
High tumor burden 
MorningLyte (NCT06284122)31  Mosunetuzumab-lenalidomide vs CIT (anti-CD20 with CHOP or benda) 790 Age, ≥18 years
FLIPI 2-5
All Ann Arbor stages
Need for treatment as per GELF 
PFS CRR
POD24
TTNT
OS
Rate of AEs
QOL outcome 
EPCORE FL-2 (NCT06191744)34  Epcoritamab-R2 vs CIT (anti-CD20 with CHOP or benda) vs R2 1080 Age, ≥18 years
FLIPI 2-5
Stage II to IV disease
Need for treatment as per GELF 
CR30 between Epco-R2 vs CIT
PFS between Epco-R2 vs CIT 
OS
PFS-2
MRD negativity rate
Outcome between Epco-R2 vs R2
QOL outcome 
OLYMPIA-1 (NCT06091254)38  Odronextamab vs CIT (R-CHOP, R-CVP, R-benda) 478 Age, ≥18 years
Stage II bulky to IV disease
Need for treatment as per GELF 
CR30 PFS
OS
TTNT
Rate of AEs
QOL outcome 
OLYMPIA-2 (NCT06097364)39  Odronextamab-CHOP/CVP vs Odronextamab-CHOP/CVP followed by odronextamab maintenance vs R-CHOP with RM 733 Age, ≥18 years
Stage II bulky to IV disease
Need for treatment as per GELF 
CR30 PFS
OS
TTNT
Rate of AEs
QOL outcome 
SOUNDTRACK-F1 (NCT06549595)40  AZD0486-rituximab vs CIT (R-CHOP, R-CVP, R-benda) 1015 Age, ≥18 years
Need for treatment as per GELF 
PFS CRR
CR30
OS 
FIL_FOLL19 (NCT05058404)41  PET CR and MRD-negative after 4 cycles CIT, complete 2 more cycles CIT vs 2 more cycles anti-CD20 only (omit chemotherapy)
All patients receive anti-CD20 maintenance 
602 Age, ≥18 years
Stage II to IV disease
Need for treatment as per GELF 
PFS OS
ORR
CRR
MRD negativity
Rate of AEs
QOL outcome 
NameTrialEnrollment targetPatientsPrimary end pointImportant secondary end points
Limited stage 
FORTplus (NCT05045664)7  RT 24 Gy + rituximab vs 4 Gy + obinutuzumab 100 Age, ≥18 years
Stage IA or IIA, nonbulky (≤7 cm)
Exclude extranodal 
CRR based on CT scan Metabolic CRR
PFS
OS
Rate of AEs 
FIL_GAZEBO (NCT05929222)8  RT 24 Gy vs 24 Gy + obintuzumab 190 Age, ≥18 years
Stage IA or IIA, or IE, nonbulky (<7 cm)
FLIPI of <2, FLIPI2 of ≤2 
PFS CRR
ORR
MRD negativity
Rate of AEs 
Low tumor burden 
SWOG2308 (NCT06337318)23  Mosunetuzumab vs rituximab 600 Age, ≥18 years
Define low tumor burden: not fulfill GELF criteria
Participants must either be experiencing distress due to disease or prefer active management rather than watch and wait
Allow patients with stage I disease who decline/are not suitable for RT 
PFS
3-year PFS 
OS
ORR
Rate of AEs 
High tumor burden 
MorningLyte (NCT06284122)31  Mosunetuzumab-lenalidomide vs CIT (anti-CD20 with CHOP or benda) 790 Age, ≥18 years
FLIPI 2-5
All Ann Arbor stages
Need for treatment as per GELF 
PFS CRR
POD24
TTNT
OS
Rate of AEs
QOL outcome 
EPCORE FL-2 (NCT06191744)34  Epcoritamab-R2 vs CIT (anti-CD20 with CHOP or benda) vs R2 1080 Age, ≥18 years
FLIPI 2-5
Stage II to IV disease
Need for treatment as per GELF 
CR30 between Epco-R2 vs CIT
PFS between Epco-R2 vs CIT 
OS
PFS-2
MRD negativity rate
Outcome between Epco-R2 vs R2
QOL outcome 
OLYMPIA-1 (NCT06091254)38  Odronextamab vs CIT (R-CHOP, R-CVP, R-benda) 478 Age, ≥18 years
Stage II bulky to IV disease
Need for treatment as per GELF 
CR30 PFS
OS
TTNT
Rate of AEs
QOL outcome 
OLYMPIA-2 (NCT06097364)39  Odronextamab-CHOP/CVP vs Odronextamab-CHOP/CVP followed by odronextamab maintenance vs R-CHOP with RM 733 Age, ≥18 years
Stage II bulky to IV disease
Need for treatment as per GELF 
CR30 PFS
OS
TTNT
Rate of AEs
QOL outcome 
SOUNDTRACK-F1 (NCT06549595)40  AZD0486-rituximab vs CIT (R-CHOP, R-CVP, R-benda) 1015 Age, ≥18 years
Need for treatment as per GELF 
PFS CRR
CR30
OS 
FIL_FOLL19 (NCT05058404)41  PET CR and MRD-negative after 4 cycles CIT, complete 2 more cycles CIT vs 2 more cycles anti-CD20 only (omit chemotherapy)
All patients receive anti-CD20 maintenance 
602 Age, ≥18 years
Stage II to IV disease
Need for treatment as per GELF 
PFS OS
ORR
CRR
MRD negativity
Rate of AEs
QOL outcome 

benda, bendamustine; CT, computed tomography; Epco, epcoritamab; FLIPI, FL International Prognostic Index; GELF, Groupe d'Etude des Lymphomes Folliculaires criteria; IE, stage 1 extranodal; ORR, overall response rate; TTNT, time to next treatment.

The GABEZO, multicenter phase 3 study (NCT05929222) is currently comparing 24 Gy RT alone against 24 Gy RT plus 8 doses of obinutuzumab for patients with limited-stage FL.8 With PFS as the primary end point, this trial aims to evaluate the benefits of adding systemic therapy. The trial will assess the potential immunosuppression toxicity from obinutuzumab, as well as the impact of an extended treatment duration on quality of life (QOL; 6 months for the combination arm compared with the 5-week course for 24 Gy RT).

Whereas the FORTplus study assesses the feasibility of reducing RT doses with a more potent CD20 mAb, the GABEZO study evaluates the effect of adding systemic therapy to RT. Both these trials will provide valuable insight on ways to limit RT toxicity, while preventing local and distant recurrence, and define the best strategies in managing limited-stage FL.

Advanced stage FL

Low tumor burden

Low tumor burden FL is defined as patients with noncontiguous stage II or advanced stages who do not meet the Groupe d'Etude des Lymphomes Folliculaires criteria. The current SOC includes watchful waiting (W&W), and rituximab monotherapy with or without short-term maintenance.9 A randomized 1:1:1 study of rituximab induction vs rituximab maintenance (RM) vs W&W in this population demonstrated longer time to next treatment in the RM group without OS benefit.10 At 10-years follow-up, 29% of patients in the W&W group remained free of treatment. Rituximab retreatment approach is supported by the RESORT study.11 Another study revealed similar response durability between short-term RM (4 weekly rituximab doses followed by 4 doses every 2 months) and long-term RM (4 weekly rituximab doses followed by every 2 months up to 5 years) supporting the use of short-term maintenance.12 These observations underscore the excellent survival experienced by most patients with low tumor burden FL and the need to prioritize safe therapies. This sets a high bar for future therapies to replace current SOC, both in terms of efficacy and toxicity considerations.

A study in early development of rituximab suggested that vitamin D (Vit-D) deficiency impaired rituximab-mediated cellular cytotoxicity and Vit-D substitution might enhance the efficacy of rituximab.13 A pooled prospective evaluation on Vit-D levels of patients with FL enrolled in clinical trials revealed that low serum Vit-D levels are associated with inferior survival outcome.14 With that in mind, ILyAD, a phase 3, randomized, double-blind, placebo-controlled study examined the combination of rituximab with Vit-D, in patients with indolent lymphoma.15 Overall, 67% of 206 patients had low tumor burden FL. Although Vit-D supplements increased serum Vit-D level without added toxicity, there was no event-free survival benefit.

NOVEL THERAPY APPROACHES

Based on the aforementioned studies, it appears that a bottleneck has been reached in terms of maximizing the use of CD20 mAb, whether through earlier treatment initiation, prolonged treatment duration, or addition of Vit-D. To address this, significant interest in the field shifted toward the use of novel therapies.

In recent years, bispecific antibodies made significant strides in the treatment of aggressive and indolent R/R B-NHL, such as diffuse large B-cell lymphoma (DLBCL) and FL.16—18 Several pivotal studies of CD3×CD20 bispecific antibodies demonstrated high efficacy in heavily pretreated FL.19—21 However, activation of T cells can lead to unique adverse events (AEs), such as cytokine release syndrome (CRS) and immune effector cell–associated neurotoxicity syndrome (ICANS).

Mosunetuzumab, a CD3×CD20 bispecific antibody, demonstrated efficacy in the third-line R/R setting, with 60% CRR and a median PFS of 24 months in its phase 2 trial for patients with FL with ≥3 previous lines.19 The phase 2 MorningSun trial (NCT05207670) supported mosunetuzumab use in the frontline setting for patients with low tumor burden FL.22 Thirty patients were enrolled and the complete metabolic response rate was 83%. Low incidences of CRS (grade 1-2, 43%) and ICANS (grade 1, 7%) were observed. To build upon this concept, SWOG 2308 (NCT06337318), a multicenter phase 3 study compared rituximab monotherapy (8 doses: 4 weekly during induction followed by 5 cycles of rituximab every 8 weeks) with mosunetuzumab (for 8 cycles).23 With PFS as the primary end point, this study aimed to challenge the concept of long-term cure by offering highly effective therapy while the tumor burden remains low but at the cost of potential added toxicity. Although mosunetuzumab is considered one of the safer bispecific antibodies, unique bispecific antibody toxicity remained. For patients with low tumor burden, it is crucial to carefully balance the benefits and risks to justify the use of more potent therapy.

High tumor burden FL

For patients with high tumor burden FL, chemoimmunotherapy (CIT) are considered the SOC at front line, as demonstrated by the BRIGHT and StiL trials.24,25 Although the RELEVANCE study revealed that rituximab-lenalidomide (R2) is as effective as CIT,26 frontline R2 is not approved by US Food and Drug Administration/European Medicines Agency, but received compendium approval based on National Comprehensive Cancer Network recommendation.27 Obinutuzumab was more efficacious over rituximab in the GALLIUM study.28 However, ∼15% to 20% of patients experience disease progression within 24 months (POD24), which is associated with higher mortality risk.29 Consequently, several ongoing frontline phase 3 clinical trials intend to address this unmet need.

A phase 1b/2 study (NCT04246086), combines mosunetuzumab with lenalidomide for untreated high disease burden FL, showed an impressive CRR of 82%.30 This is followed by the MORNINGLYTE trial (NCT06284122), which compares mosunetuzumab-lenalidomide with CIT in the frontline setting. Both arms contain maintenance anti-CD20 with total treatment duration up to 30 months.31 PFS is the primary end point. In heavily pretreated R/R FL, PFS remains a meaningful indicator of disease control. Because FL progression is heterogeneous, patients with low tumor burden relapses may be monitored with regular assessments rather than initiating immediate next-line therapy. Time to next treatment may better reflect real-world practice, but it is influenced by both physician and patient decisions regarding when to initiate subsequent therapy. The application of Groupe d'Etude des Lymphomes Folliculaires criteria remains somewhat arbitrary, highlighting the need for more standardized criteria to inform treatment initiation and retreatment decisions, potentially incorporating the use of biomarkers to improve consistency and precision. In the FL frontline setting, PFS does not necessarily correlate well with OS, as shown by StiL, GALLIUM, and PRIMA trials.25,28,32 Hence, OS end point remained important because it captures efficacy and short- and long-term toxicities of a certain treatment. As an indolent disease, newly diagnosed FL has a 10-year OS rate of >80%, necessitating long follow-up periods to detect meaningful difference in OS.32,33 Although various treatment regimens may yield similar OS outcomes, therapies that provide longer treatment-free survival may be more appealing from a patient’s perspective.

Epcoritamab, another CD3×CD20 bispecific antibody, showed efficacy with 63% CRR and 18-month PFS of 49% in its phase 2 study of third-line FL.20 EPCORE FL-2 (NCT06191744), is a phase 3 frontline trial comparing epcoritamab-R2 against CIT/R2.34 This trial has dual primary end points: PFS and complete response rate at 30 months (CR30). Although CR30 is well validated in the CIT setting as an earlier surrogacy for PFS, it may require further validation in non-CIT settings.35 Meanwhile, CR30 surrogacy was established during computed tomography scan era, but positron emission tomography (PET) scan is considered gold standard for response assessment these days.36 Secondary end points for this trial include QOL outcomes, patient reported outcomes (PRO) according to Common Terminology Criteria for Adverse Events, and minimal residual disease (MRD) negativity rate. PRO according to Common Terminology Criteria for Adverse Events allows additional dimensions to detect AEs because we know underreporting of AE from health care provider is common. Unlike the MORNINGLYTE trial, rituximab was included in the experimental arm of this trial. The study suggested that some bispecific antibodies appear combinable with CD20 mAB because they target different epitopes, do not compete for Fc-γ receptors, and induce potent target-cell killing at low occupancy rates.37 Although epcoritamab targets a different CD20 epitope (7D8) compared with rituximab, mosunetuzumab targets the same epitope (2H7).

A third CD3×CD20 bispecific antibody, odronextamab, was introduced to the FL treatment landscape based on the ELM-2 trial, which reported CRR of 73% and median PFS of 20.7 months in the third-line setting.21 The OLYMPIA-1 trial (NCT06091254) is comparing odronextamab monotherapy against CIT in frontline FL.38 CR30 is the primary end point, and multiple QOL assessments as secondary end points (Table 2). Without lenalidomide partnership in this trial, efficacy and toxicity assessment solely depends on single-agent odronextamab.

Table 2.

Details of QOL and PRO measurement included in trials

QOL assessmentComponents
EORTC QLQ-C30 5 functional scales (physical, role, cognitive, emotional, and social functioning), 3 symptom scale (fatigue, pain, and nausea/vomiting), 1 global health status/QOL scale, and 6 single items (constipation, diarrhea, insomnia, shortness of breath, appetite loss, and financial difficulties) 
FACT-Lym 15 items for NHL-related symptoms and concerns 
FACT-GP5 Single question: if participant is bothered by side effects of treatment 
EQ-5D-5L General health on 5 dimensions (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression) 
PGIC 7-Point scale depicting rating of overall change since start of treatment 
PGIS 5-Point scale depicting rating of overall severity of disease symptoms over past 7 days 
EORTC-NHL-LG20 20-Item questionnaire specifically for low-grade NHL, includes multi-item scales of symptom burden, physical condition, worries/fears, and emotional impact 
FLymSI-18 18-Item questionnaire covers 4 subscales: physical symptoms, emotional symptoms, side effects, and functional well-being 
PRO-CTCAE PRO measurement system to assess symptomatic toxicity based on frequency, severity, interference, amount, and presence/absence 
QOL assessmentComponents
EORTC QLQ-C30 5 functional scales (physical, role, cognitive, emotional, and social functioning), 3 symptom scale (fatigue, pain, and nausea/vomiting), 1 global health status/QOL scale, and 6 single items (constipation, diarrhea, insomnia, shortness of breath, appetite loss, and financial difficulties) 
FACT-Lym 15 items for NHL-related symptoms and concerns 
FACT-GP5 Single question: if participant is bothered by side effects of treatment 
EQ-5D-5L General health on 5 dimensions (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression) 
PGIC 7-Point scale depicting rating of overall change since start of treatment 
PGIS 5-Point scale depicting rating of overall severity of disease symptoms over past 7 days 
EORTC-NHL-LG20 20-Item questionnaire specifically for low-grade NHL, includes multi-item scales of symptom burden, physical condition, worries/fears, and emotional impact 
FLymSI-18 18-Item questionnaire covers 4 subscales: physical symptoms, emotional symptoms, side effects, and functional well-being 
PRO-CTCAE PRO measurement system to assess symptomatic toxicity based on frequency, severity, interference, amount, and presence/absence 

CTCAE, Common Terminology Criteria for Adverse Events; EORTC-NHL-LG20, European Organization for Research and Treatment of Cancer-Core QOL questionnaire low-grade NHL; EORTC QLQ-C30, European Organization for Research and Treatment of Cancer-Core QOL questionnaire; EQ-5D-5L, EuroQOL 5-Dimension 5-Level; FACT-GP5, Functional Assessment of Cancer Therapy, General Population; FACT-Lym, Functional Assessment of Cancer Therapy, Lymphoma; FLymSI-18, Lymphoma Symptom Index-18; PGIC, Patient Global Impression of Change; PGIS, Patient Global Impression of Severity.

The OLYMPIA-2 trial (NCT06097364) study on frontline FL therapy comprises 3 arms: odronextamab-CHOP (cyclophosphamide, doxorubicin, Oncovin [vincristine], and prednisone)/ CVP (cyclophosphamide, vincristine sulfate, and prednisone), odronextamab-CHOP/CVP with odronextamab maintenance, and rituximab-CHOP with RM.39 Primary and secondary end points are the same as in OLYMPIA-1. OLYMPIA-2 will provide insights into the role of bispecific antibody maintenance in bispecific antibody–based induction regimens.

Although plenty of treatments are focusing on targeting CD20 using mAbs and bispecific antibodies, FL tumor cells do express CD19 universally. This spurred the clinical interest in targeting both CD20 and CD19, to improve further the efficacy. AZD0486 is a novel immunoglobulin G4 (IgG4) fully human CD3×CD19 bispecific antibody that was recently reported to be effective in R/R FL (NCT04594642).57 The SOUNDTRACK-F1 (NCT06549595) frontline phase 3 trial is comparing AZD0486 plus rituximab against CIT, with PFS as the primary end point.40 Although CD20 antibodies do not appear to affect the efficacy of subsequent CD3×CD20 bispecific antibodies, first-generation CD3×CD19 bispecific antibodies blinatumomab, and B-cell maturation antigen bispecific antibodies, raised concerns regarding T-cell exhaustion for subsequent chimeric antigen receptor T-cell (CAR-T) therapy with the same target.61-63 At the same time, translational data demonstrated that, in contrast to the continuous exposure associated with blinatumomab infusion, intermittent dosing of bispecific antibodies, with treatment-free intervals, promotes T-cell expansion rather than exhaustion.63 These findings suggest that dosing strategies may significantly influence T-cell fitness and therapeutic outcomes. Future mature clinical data will be crucial to clarify the role of intermittent dosing anti-CD19 bispecific antibodies and their potential impact on the efficacy of subsequent CD19-targeted CAR-T therapies.

RESPONSE-ADAPTED CIT IN THE FRONTLINE

CIT has remained SOC in frontline FL, but tolerability can be challenging in patients who are unfit. Efforts to reduce its toxicity while maintaining efficacy through a response-adapted approach are being explored in the FIL-FOLL19 trial (NCT05058404).41 In this trial, patients receive 4 cycles of CIT induction, followed by PET scan and MRD assessment using BCL2/IgH rearrangement in the peripheral blood. Patients with PET CR and MRD negativity are randomized to either the standard arm (2 additional cycles of CIT, totaling 6) or the experimental arm, which omits further CIT. The primary end point is PFS, with QOL as one of the secondary end points. This trial recalls the FOLL12 study, which used a response-adapted approach to determine postinduction treatment strategies.64 In FOLL12, the response-adapted arm showed an inferior PFS compared with the standard RM arm, with 3-year PFS of 72% vs 86%, respectively (P < .001).

As previously highlighted in the FOLL12 study, the practicality of using peripheral blood BCL2/IgH rearrangement as MRD assessment in the FOLL19 trial poses challenges, including technical limitations and lower sensitivity. These factors may limit the applicability of this approach in real-world settings. Conversely, circulating tumor DNA (ctDNA) offers a promising alternative, with the potential to overcome these limitations through improved sensitivity and broader clinical feasibility.

ctDNA, also known as tumor-derived cell-free DNA, is released into the peripheral blood by tumor cells after biological processes of apoptosis, proliferation, and necrosis. Next-generation sequencing–based immunoglobulin sequencing, which identifies and tracks unique disease-associated immunoglobulin sequences (IgH, IgK, and IgL) in blood or bone marrow samples, best known as clonoSEQ commercially, is currently being explored in various B-cell malignancies.65-67 This method proved highly tumor specific, with a low background error rate but limited by the total input DNA. This impairs the performance of the assay when ctDNA levels are low. At a single point at end of treatment (EOT), the sensitivity to detect relapses was ∼30%.67 

In contrast, a different MRD detection platform using the phased variant enrichment and detection sequencing (PhaseED-seq), has shown utility in detecting and quantifying ctDNA at levels as low as 1 part per million.68 This method takes advantage the proximity of single-nucleotide variants in the BCL genome, and detects and tracks multiple somatic variants located on the same cell-free DNA molecule. This resulted in higher sensitivity, translated to more reliable detection of EOT MRD of up to 80% to 90%.69,70 Several studies in DLBCL have demonstrated that PhaseED-seq correlates strongly with EOT disease status, offering superior sensitivity and specificity compared with PET scans.70-72 Early data suggest that ctDNA could also be useful in the field of FL, predict time to first treatment in untreated patients, and assess treatment response and outcomes.73,74 However, data on MRD in FL using either clonoSEQ or PhaseED-seq in FL remains limited compared with DLBCL. The recognition and understanding of ctDNA in DLBCL has improved over years, leading to its recommendation by the National Comprehensive Cancer Network guidelines as an EOT assessment modality for patients with DLBCL with positive PET scans, when a biopsy is not feasible.27 

Summary on frontline trials

Standing at the forefront of these exciting frontline phase 3 trials, there is hope that we may finally answer the longstanding question of curing FL, without compromising toxicity and QOL. Several important questions remain to be addressed: can we treat without chemotherapy? Are rituximab or lenalidomide essential partners for CD3×CD20 bispecific antibodies? What is the role of maintenance in bispecific antibody therapy? Can we revive response-adapted treatment approaches in FL?

CAR-T therapies

Autologous anti-CD19 CAR-T therapies have been a breakthrough for B-NHL, including FL. These genetically engineered T cells recognize CD19 on B cells and initiate cytotoxic antitumor activity independent of major histocompatibility complex (MHC) presentation.75 Although this MHC independence is a key advantage, it also limits CAR-T ability to recognize intracellular tumor-associated antigen, which require MHC-mediated presentation.

Three CAR-T products, axicabtagene ciloleucel (axi-cel), tisagenlecleucel (tisa-cel), and lisocabtagene maraleucel (liso-cel), are approved for R/R FL in the third-line setting. Axi-cel, which uses a CD28 costimulatory domain, showed high efficacy in the ZUMA-5 trial (FL cohort: CRR, 79%; median PFS, 57 months) but with notable toxicity (grade ≥3 CRS, 6%; grade ≥3 ICANS, 15%). Tisa-cel and liso-cel, both using 4-1BB domains, demonstrated strong efficacy with lower toxicity. In the ELARA trial (median 4 previous lines), tisa-cel achieved a CRR of 68% and 3-year PFS of 53%, with no grade ≥3 CRS and only 1% ICANS. In TRANSCEND FL (≥2 previous lines), liso-cel reported a CRR of 94% and 2-year PFS of 73%, with 1% grade ≥3 CRS and 2% ICANS. These results support the growing role of CAR-T therapy in R/R FL, with ongoing trials aiming to further optimize outcomes Table 3.

Table 3.

Features and outcome of selected trials with reported outcomes

TrialPhaseNPatientsRisk groupCR rate (%)1-year PFS (%)
ZUMA-5, axi-cel51  124 R/R Median previous line 3
POD24, 56% 
79 74 
ELARA, tisa-cel52  97 R/R Median previous line 4
POD24, 63% 
68 67 
TRANSCEND FL, liso-cel53  101 (3L+ cohort) R/R Median previous line 3
POD24, 54% 
94 81 
Mosunetuzumab19  90 R/R Median previous line 3
POD24, 52% 
60 58 
Epcoritamab20  1/2 128 R/R Median previous line 3
POD24, 42% 
63 18-month PFS, 49 
Odronextamab21  128 R/R Median previous line 3
POD24, 49% 
73 66 
InMIND, tafa-R2 vs R254  548 R/R Median previous line 1
POD24, 32% 
52 (tafa-R2)
41 (R2) 
Median,
22.4 months (tafa-R2)
13.9 months (R2) 
Mosunetuzumab-lenalidomide55  1b 27 R/R Median previous line 1
POD24, 11% 
77 Not reported 
Epcoritamab-R256  1/2 111 R/R Median previous line 1
POD24, 38% 
87 21-month PFS, 80 
AZD048657  56 R/R Median previous line 3
POD24, 34% 
85 79 
Tazemetostat58  99 R/R Median previous line 2-3
POD24, 42%-59% (based on EZH2 mutation status) 
13 (EZH2 mutated), 4 (EZH2 wild type) Median, 11-14 months 
ROSEWOOD, ZO49  145 R/R Median previous line 3
POD24, 34% 
39 Median, 28 months 
Golcadomide with/without rituximab59  1/2 63 R/R Median previous line 4 (golcadomide monotherapy)
Median previous line 3 (golcadomide-rituximab) 
42 (golcadomide monotherapy)
67 (golcadomide 0.4 mg + rituximab) 
Median, 22 months (golcadomide monotherapy) 
Iberdomide with obinutuzumab60  1/2 13 (FL + MZL cohort) R/R Median previous line 4 (all B-NHL) 39 (FL + MZL) Not reported 
MORNINGSUN, mosunetuzumab22  30 Front line, low tumor burden FLIPI 3-5, 10% 83 Not reported 
Mosunetuzumab-lenalidomide30  1b/2 37 Front line, high tumor burden Stage III-IV, 87%
FLIPI 3-5, 46% 
82 Not reported 
TrialPhaseNPatientsRisk groupCR rate (%)1-year PFS (%)
ZUMA-5, axi-cel51  124 R/R Median previous line 3
POD24, 56% 
79 74 
ELARA, tisa-cel52  97 R/R Median previous line 4
POD24, 63% 
68 67 
TRANSCEND FL, liso-cel53  101 (3L+ cohort) R/R Median previous line 3
POD24, 54% 
94 81 
Mosunetuzumab19  90 R/R Median previous line 3
POD24, 52% 
60 58 
Epcoritamab20  1/2 128 R/R Median previous line 3
POD24, 42% 
63 18-month PFS, 49 
Odronextamab21  128 R/R Median previous line 3
POD24, 49% 
73 66 
InMIND, tafa-R2 vs R254  548 R/R Median previous line 1
POD24, 32% 
52 (tafa-R2)
41 (R2) 
Median,
22.4 months (tafa-R2)
13.9 months (R2) 
Mosunetuzumab-lenalidomide55  1b 27 R/R Median previous line 1
POD24, 11% 
77 Not reported 
Epcoritamab-R256  1/2 111 R/R Median previous line 1
POD24, 38% 
87 21-month PFS, 80 
AZD048657  56 R/R Median previous line 3
POD24, 34% 
85 79 
Tazemetostat58  99 R/R Median previous line 2-3
POD24, 42%-59% (based on EZH2 mutation status) 
13 (EZH2 mutated), 4 (EZH2 wild type) Median, 11-14 months 
ROSEWOOD, ZO49  145 R/R Median previous line 3
POD24, 34% 
39 Median, 28 months 
Golcadomide with/without rituximab59  1/2 63 R/R Median previous line 4 (golcadomide monotherapy)
Median previous line 3 (golcadomide-rituximab) 
42 (golcadomide monotherapy)
67 (golcadomide 0.4 mg + rituximab) 
Median, 22 months (golcadomide monotherapy) 
Iberdomide with obinutuzumab60  1/2 13 (FL + MZL cohort) R/R Median previous line 4 (all B-NHL) 39 (FL + MZL) Not reported 
MORNINGSUN, mosunetuzumab22  30 Front line, low tumor burden FLIPI 3-5, 10% 83 Not reported 
Mosunetuzumab-lenalidomide30  1b/2 37 Front line, high tumor burden Stage III-IV, 87%
FLIPI 3-5, 46% 
82 Not reported 

3L+, third-line and beyond; tafa, tafasitamab.

The ZUMA-22 trial (NCT05371093) is an ongoing phase 3 study examining axi-cel in comparison with SOC for patients with R/R FL who have had at least 2 previous lines of treatment.42 Notably, this study permits the inclusion of patients with POD24 in the second-line setting. Primary end point is PFS, with secondary end points including various QOL assessments. Importantly, this study incorporate transition into a long-term follow-up study lasting up to 15 years from randomization, which will provide valuable insights into the long-term effects of CAR-T therapy Table 4.

Table 4.

Features of ongoing phase 3 R/R FL trials

NameTrialEnrollment targetPatientsPrimary end pointImportant secondary end points
ZUMA-22 (NCT05371093)42  Axi-cel vs SOC (R2, R-CHOP, R-benda) 230 Age, ≥18 years
R/R after first-line CIT with POD24
After ≥2 previous lines systemic therapy 
PFS OS
CRR
ORR
TTNT
Rate of AEs
QOL outcome 
LEDA (NCT05888493)43  Tisa-cel vs SOC (R2, R-CHOP) 108 Age, ≥18 years
R/R after ≥2 previous lines systemic therapy 
PFS OS
CRR
ORR
TTNT
Rate of AEs 
TRANSFORM FL (NCT06313996)44  Liso-cel vs SOC (R2, R-CHOP, R-benda) 300 Age, ≥18 years
R/R after 2-3 previous lines including anti-CD20 and alkylator
R/R after 1 previous line including anti-CD20 and alkylator with high-risk features 
PFS OS
CRR
ORR
TTNT
PFS-2
Rate of AEs
QOL outcome 
CELESTIMO (NCT04712097)45  Mosunetuzumab-lenalidomide vs R2 474 Age, ≥18 years
R/R after ≥1 previous lines systemic therapy including immunotherapy or CIT 
PFS OS
CRR
ORR
TTNT
Rate of AEs
QOL outcome 
EPCORE FL-1 (NCT05409066)46  Epcoritamab-R2 vs R2 500 Age, ≥18 years
R/R after ≥1 previous lines of CIT (exclude anti-CD20 monotherapy) 
PFS OS
CRR
MRD negativity 
OLYMPIA-5 (NCT06149286)47  Odronextamab-lenalidomide vs R2 470 (FL and MZL) Age, ≥18 years
R/R after ≥1 previous lines systemic therapy including immunotherapy or CIT 
PFS OS
CRR
TTNT
Rate of AEs
QOL outcome 
SYMPHONY-1 (NCT04224493)48  R2-tazemetostat vs R2-placebo 612 Age, ≥18 years
R/R after ≥1 previous lines systemic therapy including immunotherapy or CIT or chemotherapy
No restriction on EZH2 mutation status 
PFS OS
CRR
ORR
TTNT
Rate of AEs
QOL outcome 
MAHOGANY (NCT05100862)49  ZO vs R2 750 (FL and MZL) Age, ≥18 years
R/R after ≥1 previous lines systemic therapy including anti-CD20 
PFS OS
CRR
ORR
TTNT
Rate of AEs
QOL outcome 
GOLSEEK-4 (NCT06911502)50  Golcadomide-rituximab vs investigator’s choice (R2/CIT) 400 Age, ≥18 years
R/R after ≥1 previous lines systemic therapy including anti-CD20 and an alkylating agent 
PFS OS
ORR
TTNT
PFS-2
QOL outcome
MRD negativity 
NameTrialEnrollment targetPatientsPrimary end pointImportant secondary end points
ZUMA-22 (NCT05371093)42  Axi-cel vs SOC (R2, R-CHOP, R-benda) 230 Age, ≥18 years
R/R after first-line CIT with POD24
After ≥2 previous lines systemic therapy 
PFS OS
CRR
ORR
TTNT
Rate of AEs
QOL outcome 
LEDA (NCT05888493)43  Tisa-cel vs SOC (R2, R-CHOP) 108 Age, ≥18 years
R/R after ≥2 previous lines systemic therapy 
PFS OS
CRR
ORR
TTNT
Rate of AEs 
TRANSFORM FL (NCT06313996)44  Liso-cel vs SOC (R2, R-CHOP, R-benda) 300 Age, ≥18 years
R/R after 2-3 previous lines including anti-CD20 and alkylator
R/R after 1 previous line including anti-CD20 and alkylator with high-risk features 
PFS OS
CRR
ORR
TTNT
PFS-2
Rate of AEs
QOL outcome 
CELESTIMO (NCT04712097)45  Mosunetuzumab-lenalidomide vs R2 474 Age, ≥18 years
R/R after ≥1 previous lines systemic therapy including immunotherapy or CIT 
PFS OS
CRR
ORR
TTNT
Rate of AEs
QOL outcome 
EPCORE FL-1 (NCT05409066)46  Epcoritamab-R2 vs R2 500 Age, ≥18 years
R/R after ≥1 previous lines of CIT (exclude anti-CD20 monotherapy) 
PFS OS
CRR
MRD negativity 
OLYMPIA-5 (NCT06149286)47  Odronextamab-lenalidomide vs R2 470 (FL and MZL) Age, ≥18 years
R/R after ≥1 previous lines systemic therapy including immunotherapy or CIT 
PFS OS
CRR
TTNT
Rate of AEs
QOL outcome 
SYMPHONY-1 (NCT04224493)48  R2-tazemetostat vs R2-placebo 612 Age, ≥18 years
R/R after ≥1 previous lines systemic therapy including immunotherapy or CIT or chemotherapy
No restriction on EZH2 mutation status 
PFS OS
CRR
ORR
TTNT
Rate of AEs
QOL outcome 
MAHOGANY (NCT05100862)49  ZO vs R2 750 (FL and MZL) Age, ≥18 years
R/R after ≥1 previous lines systemic therapy including anti-CD20 
PFS OS
CRR
ORR
TTNT
Rate of AEs
QOL outcome 
GOLSEEK-4 (NCT06911502)50  Golcadomide-rituximab vs investigator’s choice (R2/CIT) 400 Age, ≥18 years
R/R after ≥1 previous lines systemic therapy including anti-CD20 and an alkylating agent 
PFS OS
ORR
TTNT
PFS-2
QOL outcome
MRD negativity 

MZL, marginal zone lymphoma.

For tisa-cel, the LEDA trial (NCT05888493), building on the findings of the ELARA study, is comparing tisa-cel with SOC for patients with FL with at least 2 previous lines of treatment.43 Unlike ZUMA-22, this trial does not include a separate cohort for patients with POD24 in the second-line setting, which may influence the clinical positioning of tisa-cel, particularly if axi-cel gains approval for use in this high-risk subgroup.

The planned phase 3 TRANSFORM FL trial (NCT06313996) will compare liso-cel with SOC in the second-line setting, with PFS as the primary end point, and PFS-2 (defined as the time from start of current therapy to disease progression or death on next line of therapy) among the secondary end points.44 PFS-2 is an interesting end point, because it assesses the impact of second-line anti-CD19 CAR-T therapy on subsequent lines of treatment. Although all the aforementioned anti-CD19 CAR-T studies allow SOC relapsed regimens as control arms, these are limited to either Bendamustine-Rituximab (BR)/R-CHOP or R2 therapy.

Bispecific antibodies

Although CD3×CD20 bispecific antibodies are being explored in first-line trials, there are ongoing phase 3 trials evaluating their use in the R/R setting. The CELESTIMO trial (NCT04712097) compares mosunetuzumab-lenalidomide with R2 in the second-line setting.45 The primary end point is PFS. Practical application of this trial may be influenced by results from frontline bispecific antibody trials. Similarly, the EPCORE FL-1 trial (NCT05409066) is investigating epcoritamab plus R2 against R2 in second line.46 This trial adopts a finite epcoritamab treatment duration, with maximum 12 cycles of epcoritamab-R2. This provides insight into the durability and feasibility of retreatment, as well as facilitate comparison with other bispecific antibodies. Finally, the OLYMPIA-5 trial (NCT06149286) is a phase 3 study comparing odronextamab-lenalidomide with R2 in the second line, with PFS as the primary end point, with multiple QOL secondary end points.47 It will be interesting to see how these 3 CD3×CD20 bispecific antibodies challenge the current second-line space, which is predominantly occupied by R2.

Targeted therapies

Because of its epigenetic alterations, FL often harbors enhancer of Zeste homolog (EZH2) mutations.76 Tazemetostat, an EZH2 inhibitor, is approved for use in R/R FL, showing efficacy in both EZH2-mutated and wild-type patients.58 The overall response rate ranges from 35% to 69%, with median PFS of ∼11 to 14 months. Tazemetostat is an oral therapy with favorable toxicity profile. A phase 3 trial, SYMPHONY-1 (NCT04224493), is comparing R2-tazemetostat with R2-placebo.48 Tazemetostat or placebo is administered for 2 years, with PFS as the primary end point. This study, by including a placebo arm, provides valuable insights into the toxicity differences between doublet and triplet combinations. It also separates patients into cohorts based on their EZH2 mutation status, allowing for a more granular understanding of efficacy.

In the ROSEWOOD study, obinutuzumab and zanubrutinib (ZO) offered a synergistic effect in R/R FL.49 The MAHOGANY trial (NCT05100862) is a phase 3 study comparing ZO with R2 in patients with R/R FL.77 Zanubrutinib is administered continuously until progression, whereas lenalidomide in the R2 arm is limited to 12 cycles. PFS is the primary end point, with QOL measurement among the secondary end points. It is important to consider the differences in treatment duration and anti-CD20 partner when interpreting the results. Nevertheless, ZO may be an attractive option for patients who are unfit or frail, given its favorable oral administration route after obinutuzumab induction.

CELMods

Unlike traditional immunomodulatory drugs, cereblon E3 ligase modulators (CELMods) have been designed to have improved potency and selectivity, leading to greater antitumor activity and lesser side effects. Several CELMods, such as golcadomide and iberdomide, are now being investigated in several B-cell malignancies.59,60,78 GOLSEEK-4 (NCT06911502) is a phase 3 trial that compares the efficacy and safety of golcadomide in combination with rituximab against investigator choice (R2 or CIT) in patients with R/R FL who received at least 1 previous line of CD20 mAb and alkylating agent.50 The primary end point is PFS, with MRD data and QOL assessment among the secondary end points. In contrast to the other aforementioned R/R FL phase 3 bispecific antibodies and targeted therapies, this study aims to question the role of lenalidomide in the second line and beyond. Lenalidomide is known to be myelosuppressive and prothrombotic, with potential dermatologic and gastrointestinal side effects, which sometimes affect patient QOL and tolerability.

The AUGMENT study established the important role of 12 cycles of R2 as a second-line treatment for R/R FL.79 Recently, the inMIND study (NCT04680052) demonstrated that the addition of tafasitamab, a CD19 mAb, to R2 significantly improved PFS compared with R2 alone in patients with R/R FL.54 Notably, R2 has become a common control arm in several ongoing phase 3 trials for relapsed disease, including studies evaluating bispecific antibodies, tazemetostat, and zanubrutinib. This consistent use of R2 as a comparator provides a valuable benchmark for cross-trial comparisons, enabling better assessment of the relative efficacy of emerging therapies.

Unlike CELESTIMO and OLYMPIA-5, the EPCORE FL-1 trial excludes anti-CD20 monotherapy as a previous line of treatment. This means that all enrolled patients have had at least 1 line of anti-CD20 in combination with chemotherapy, reflecting a potentially higher-risk cohort, including those who experienced POD24 after CIT.80 

Several large-scale trials, such as SYMPHONY-1 and CELESTIMO, have rapidly advanced from small phase 1 studies to large, randomized phase 3 trials. Although this accelerated development is welcomed, there is a concern that such rapid progression may overlook or underestimate potential toxicities, particularly the less well-characterized immune-related AEs associated with bispecific antibodies.

Earlier use of CAR-Ts in DLBCL allows better T-cell fitness with promising outcome (eg, ZUMA-12, ZUMA-7).81,82 However, in FL, this approach may result in a shorter time interval between frontline bendamustine exposure and CAR-T apheresis. A study indicated that bendamustine exposure <9 months before CAR-T apheresis had a detrimental effect on CAR-T efficacy.83 Although the impact of previous bendamustine on bispecific antibody efficacy is less pronounced, this is based on limited number of retrospective cases.84,85 As these trials unfold, we will gain valuable insights into the best strategies for positioning CAR-Ts in FL.

With insights gained from the ongoing trials, there is growing potential to better define the optimal second-line treatment for high-risk patients with R/R FL, particularly those experiencing POD24. Although POD24 is widely recognized as a high-risk event, its current definition has limitations; it may encompass patients with transformed LBCL as well as those with persistent indolent FL. A study demonstrated differences in survival outcome based on histology at the time of progression.86 An added layer of heterogeneity stems from the variable feasibility of obtaining biopsies from the most suspicious sites at each relapse. These challenges contribute to variability in baseline risk among patients with POD24 enrolled in R/R FL trials, potentially confounding the interpretation of outcomes. As such, there is an urgent need for more precise a priori biomarkers to enhance risk stratification and guide individualized treatment selection.

Although we are excited and eager to hear the outcomes of these trials, it is important to note that their results may not be directly applicable to patients who are frail or those who are unfit for CIT or lenalidomide as part of the control arms. For older adults, incorporating geriatric assessments into the inclusion criteria could help address this issue, providing a more standardized and objective approach to patient recruitment.87 

Given the long and evolving course of FL, incorporating patients’ values and preferences is essential for shared decision-making.88 A qualitative study found that patients with FL prefer a holistic, individualized approach that accounts for lifestyle, side effect concerns, and treatment burden.89 Other studies highlight that physicians and patients may prioritize different factors: while both value life expectancy and disease control, patients are often more willing to trade some disease control for simpler administration, lower toxicity, and fewer monitoring visits.90,91 As efficacy differences between active therapies narrow, PROs related to toxicity and QOL are becoming increasingly important. Going forward, the key differentiator in treatment selection may be the option that offers the lowest toxicity and best QOL outcomes.92 Unfortunately, there is currently a lack of standardization in QOL assessments across various clinical trials particularly FL-specific QOL scale, even within the same therapeutic area. This complicates meaningful comparisons between treatment arms using disease-specific measures.

Lastly, although CAR-Ts, bispecific antibodies, and targeted therapies are poised to affect the future treatment landscape of lymphoma, health care disparities remain a critical challenge. This includes practice variations between community-based and academic centers, and low- to middle-income countries’ accessibility to novel therapies. Given the rising costs of novel therapies, well-designed cost-effectiveness studies are essential to justify their use in earlier-line treatments.

With numerous antilymphoma strategies being tested in ongoing phase 3 trials, the future of FL treatment is exciting. We hope to establish a refined SOC that meets the diverse needs of patients, including the potential for a cure, minimizing toxicity, and maintaining a high QOL. As science often unfolds, the results from these trials will likely raise new questions. Careful methodological evaluation and thoughtful discussion will be crucial in guiding future studies, enabling the practice of personalized medicine tailored to each patient’s needs.

Figures on graphical abstract were created with BioRender.com.

Contribution: L.C.K.N and C.C designed the research, summarized the data, and wrote and approved the manuscript.

Conflict-of-interest disclosure: C.C. reports consultancy with, and have received honoraria from Bristol Myers Squibb, AbbVie, Genentech/Roche, and Genmab; received research funding from Genmab, Gilead, and Genentech; and reports a leadership role with the American Society of Hematology and the Lymphoma Research Foundation. L.C.K.N. declares no competing financial interests.

Correspondence: Carla Casulo, Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Box 704, Rochester, NY 14642; email: carla_casulo@urmc.rochester.edu.

1.
Teras
LR
,
DeSantis
CE
,
Cerhan
JR
,
Morton
LM
,
Jemal
A
,
Flowers
CR
.
2016 US lymphoid malignancy statistics by World Health Organization subtypes
.
CA Cancer J Clin
.
2016
;
66
(
6
):
443
-
459
.
2.
National Cancer Institute
.
Cancer stat facts: NHL-follicular lymphoma
. Accessed 19 February 2025. https://seer.cancer.gov/statfacts/html/follicular.html.
3.
Sarkozy
C
,
Maurer
MJ
,
Link
BK
, et al
.
Cause of death in follicular lymphoma in the first decade of the rituximab era: a pooled analysis of French and US cohorts
.
J Clin Oncol
.
2019
;
37
(
2
):
144
-
152
.
4.
Rivas-Delgado
A
,
Magnano
L
,
Moreno-Velázquez
M
, et al
.
Response duration and survival shorten after each relapse in patients with follicular lymphoma treated in the rituximab era
.
Br J Haematol
.
2019
;
184
(
5
):
753
-
759
.
5.
Brady
JL
,
Binkley
MS
,
Hajj
C
, et al
.
Definitive radiotherapy for localized follicular lymphoma staged by 18F-FDG PET-CT: a collaborative study by ILROG
.
Blood
.
2019
;
133
(
3
):
237
-
245
.
6.
Hoskin
P
,
Popova
B
,
Schofield
O
, et al
.
4 Gy versus 24 Gy radiotherapy for follicular and marginal zone lymphoma (FoRT): long-term follow-up of a multicentre, randomised, phase 3, non-inferiority trial
.
Lancet Oncol
.
2021
;
22
(
3
):
332
-
340
.
7.
Early stage follicular lymphoma and radio therapy PLUS anti-CD20 antibody (FORTplus). ClinicalTrials.gov identifier: NCT05045664
. Updated 06 December 2024. Accessed 8 January 2025. https://clinicaltrials.gov/study/NCT05045664.
8.
Comparison between local radiotherapy alone or combined with obinutuzumab in early stage follicular lymphoma: the GAZEBO trial from the Fondazione Italiana Linfomi (FIL_GAZEBO). ClinicalTrials.gov identifier: NCT05929222
. Updated 24 December 2024. Accessed 8 January 2025. https://clinicaltrials.gov/study/NCT05929222.
9.
Cartron
G
,
Trotman
J
.
Time for an individualized approach to first-line management of follicular lymphoma
.
Haematologica
.
2022
;
107
(
1
):
7
-
18
.
10.
Northend
M
,
Wilson
W
,
Clifton-Hadley
L
, et al
.
Long term follow-up of international randomised phase 3 study of rituximab versus a watch and wait approach for patients with asymptomatic, low tumour burden follicular lymphoma shows rituximab is highly effective at delaying time to new treatment without detrimental impact following next line of therapy [abstract]
.
Blood
.
2022
;
140
(
suppl 1
):
1456
-
1458
.
11.
Kahl
BS
,
Jegede
OA
,
Peterson
C
, et al
.
Long-term follow-up of the RESORT study (E4402): a randomized phase III comparison of two different rituximab dosing strategies for low-tumor burden follicular lymphoma
.
J Clin Oncol
.
2024
;
42
(
7
):
774
-
778
.
12.
Moccia
AA
,
Taverna
C
,
Schär
S
, et al
.
Prolonged rituximab maintenance in follicular lymphoma patients: long-term results of the SAKK 35/03 randomized trial
.
Blood Adv
.
2020
;
4
(
23
):
5951
-
5957
.
13.
Bittenbring
JT
,
Neumann
F
,
Altmann
B
, et al
.
Vitamin D deficiency impairs rituximab-mediated cellular cytotoxicity and outcome of patients with diffuse large B-cell lymphoma treated with but not without rituximab
.
J Clin Oncol
.
2014
;
32
(
29
):
3242
-
3248
.
14.
Kelly
JL
,
Salles
G
,
Goldman
B
, et al
.
Low serum vitamin D levels are associated with inferior survival in follicular lymphoma: a prospective evaluation in SWOG and LYSA studies
.
J Clin Oncol
.
2015
;
33
(
13
):
1482
-
1490
.
15.
Friedberg
JW
,
Brady
MT
,
Strawderman
M
, et al
.
Vitamin D in patients with low tumor-burden indolent non-Hodgkin lymphoma treated with rituximab therapy (ILyAD): a randomized, phase 3 clinical trial
.
EClinicalMedicine
.
2024
;
78
:
102959
.
16.
Hutchings
M
,
Morschhauser
F
,
Iacoboni
G
, et al
.
Glofitamab, a novel, bivalent CD20-targeting T-cell-engaging bispecific antibody, induces durable complete remissions in relapsed or refractory B-cell lymphoma: a phase I trial
.
J Clin Oncol
.
2021
;
39
(
18
):
1959
-
1970
.
17.
van der Horst
HJ
,
de Jonge
AV
,
Hiemstra
IH
, et al
.
Epcoritamab induces potent anti-tumor activity against malignant B-cells from patients with DLBCL, FL and MCL, irrespective of prior CD20 monoclonal antibody treatment
.
Blood Cancer J
.
2021
;
11
(
2
):
38
.
18.
Budde
LE
,
Assouline
S
,
Sehn
LH
, et al
.
Single-agent mosunetuzumab shows durable complete responses in patients with relapsed or refractory B-cell lymphomas: phase I dose-escalation study
.
J Clin Oncol
.
2022
;
40
(
5
):
481
-
491
.
19.
Sehn
LH
,
Bartlett
NL
,
Matasar
MJ
, et al
.
Long-term 3-year follow-up of mosunetuzumab in relapsed or refractory follicular lymphoma after ≥2 prior therapies
.
Blood
.
2025
;
145
(
7
):
708
-
719
.
20.
Linton
KM
,
Vitolo
U
,
Jurczak
W
, et al
.
Epcoritamab monotherapy in patients with relapsed or refractory follicular lymphoma (EPCORE NHL-1): a phase 2 cohort of a single-arm, multicentre study
.
Lancet Haematol
.
2024
;
11
(
8
):
e593
-
e605
.
21.
Kim
TM
,
Taszner
M
,
Novelli
S
, et al
.
Safety and efficacy of odronextamab in patients with relapsed or refractory follicular lymphoma
.
Ann Oncol
.
2024
;
35
(
11
):
1039
-
1047
.
22.
Flinn
IW
,
Budde
LE
,
Tun
AM
, et al
.
Subcutaneous mosunetuzumab is active and has a manageable safety profile in patients with previously untreated, low-tumor burden follicular lymphoma: initial results from the phase II morningsun study [abstract]
.
Blood
.
2023
;
142
(
suppl 1
):
3029
.
23.
Comparing rituximab and mosunetuzumab drug treatments for people with low tumor burden follicular lymphoma (SWOG 2308). ClinicalTrials.gov identifier: NCT06337318
. Updated 8 January 2025. Accessed 8 January 2025. https://clinicaltrials.gov/study/NCT06337318.
24.
Flinn
IW
,
van der Jagt
R
,
Kahl
BS
, et al
.
Randomized trial of bendamustine-rituximab or R-CHOP/R-CVP in first-line treatment of indolent NHL or MCL: The BRIGHT study
.
Blood
.
2014
;
123
(
19
):
2944
-
2952
.
25.
Rummel
MJ
,
Niederle
N
,
Maschmeyer
G
, et al;
Study group indolent Lymphomas (StiL)
.
Bendamustine plus rituximab versus CHOP plus rituximab as first-line treatment for patients with indolent and mantle-cell lymphomas: an open-label, multicentre, randomised, phase 3 non-inferiority trial
.
Lancet
.
2013
;
381
(
9873
):
1203
-
1210
.
26.
Morschhauser
F
,
Nastoupil
L
,
Feugier
P
, et al
.
Six-year results from RELEVANCE: lenalidomide plus rituximab (R2) versus rituximab-chemotherapy followed by rituximab maintenance in untreated advanced follicular lymphoma
.
J Clin Oncol
.
2022
;
40
(
28
):
3239
-
3245
.
27.
National Comprehensive Cancer Network
.
NCCN clinical practice guidelines in oncology (NCCN guidelines®) for B-cell lymphoma version 2.2025
. 2025. Accessed 20 February 2025. https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1480.
28.
Townsend
W
,
Hiddemann
W
,
Buske
C
, et al
.
Obinutuzumab versus rituximab immunochemotherapy in previously untreated iNHL: final results from the GALLIUM study
.
Hemasphere
.
2023
;
7
(
7
):
e919
.
29.
Casulo
C
,
Dixon
JG
,
Le-Rademacher
J
, et al
.
Validation of POD24 as a robust early clinical end point of poor survival in FL from 5225 patients on 13 clinical trials
.
Blood
.
2022
;
139
(
11
):
1684
-
1693
.
30.
Morschhauser
F
,
Patel
K
,
Bobillo
S
, et al
.
Preliminary findings of a phase Ib/II trial indicate manageable safety and promising efficacy for mosunetuzumab in combination with lenalidomide (M+Len) in previously untreated (1L) follicular lymphoma (FL) [abstract]
.
Blood
.
2023
;
142
(
suppl 1
):
605
.
31.
Study of mosunetuzumab plus lenalidomide compared to anti-CD20 anti-body + chemotherapy in follicular lymphoma FLIPI2-5 (MorningLyte). ClinicalTrials.gov identifier: NCT06284122
. Updated 15 October 2024. Accessed 8 January 2025. https://clinicaltrials.gov/study/NCT06284122.
32.
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
.
33.
Liu
Q
,
Fayad
L
,
Cabanillas
F
, et al
.
Improvement of overall and failure-free survival in stage IV follicular lymphoma: 25 years of treatment experience at The University of Texas M.D. Anderson Cancer Center
.
J Clin Oncol
.
2006
;
24
(
10
):
1582
-
1589
.
34.
Linton
KM
,
Thompson
P
,
Luminari
S
, et al
.
EPCORE FL-2: Phase 3 trial of epcoritamab with rituximab and lenalidomide (R2) vs chemoimmunotherapy or R2 in previously untreated follicular lymphoma [abstract]
.
J Clin Oncol
.
2024
;
42
(
suppl 19
):
TPS7084
.
35.
Shi
Q
,
Flowers
CR
,
Hiddemann
W
, et al
.
Thirty-month complete response as a surrogate end point in first-line follicular lymphoma therapy: an individual patient-level analysis of multiple randomized trials
.
J Clin Oncol
.
2017
;
35
(
5
):
552
-
560
.
36.
Bachy
E
,
Rufibach
K
,
Parreira
J
,
Launonen
A
,
Nielsen
T
,
Hackshaw
A
.
Phase III clinical trials in first-line follicular lymphoma: a review of their design and interpretation
.
Adv Ther
.
2021
;
38
(
7
):
3489
-
3505
.
37.
Falchi
L
,
Vardhana
SA
,
Salles
GA
.
Bispecific antibodies for the treatment of B-cell lymphoma: promises, unknowns, and opportunities
.
Blood
.
2023
;
141
(
5
):
467
-
480
.
38.
Birhiray
RE
,
Luminari
S
,
Kim
TM
, et al
.
Phase 3 trial evaluating the efficacy and safety of odronextamab versus investigator’s choice in previously untreated follicular lymphoma (OLYMPIA-1) [abstract]
.
J Clin Oncol
.
2024
;
42
(
suppl 16
):
TPS7096
.
39.
Hardin
C
,
Gray
C
,
Novelli
S
, et al
.
Phase 3 trial evaluating the efficacy and safety of odronextamab plus chemotherapy versus rituximab plus chemotherapy in previously untreated follicular lymphoma (OLYMPIA-2) [abstract]
.
J Clin Oncol
.
2024
;
42
(
suppl 16
):
TPS7099
.
40.
A study of AZD0486 plus rituximab in previously untreated follicular lymphoma patients. ClinicalTrials.gov identifier: NCT06549595
. Updated 18 December 2024. Accessed 8 January 2025. https://clinicaltrials.gov/study/NCT06549595.
41.
Shortened vs standard chemotherapy combined with immunotherapy for the initial treatment of patients with high tumor burden follicular lymphoma (FIL_FOLL19). ClinicalTrials.gov identifier: NCT05058404
. Updated 10 July 2024. Accessed 8 January 2025. https://clinicaltrials.gov/study/NCT05058404.
42.
Flinn
IW
,
Jacobson
CA
,
Nastoupil
LJ
, et al
.
ZUMA-22: A phase 3, randomized controlled study of axicabtagene ciloleucel (axi-cel) versus standard-of-care therapy in patients with relapsed or refractory (R/R) follicular lymphoma (FL) [abstract]
.
Journal of Clinical Oncology
.
2023
;
41
(
suppl 16
):
TPS7579
.
43.
A phase III trial comparing tisagenlecleucel to standard of care (SoC) in adult participants with R/R follicular lymphoma (LEDA). ClinicalTrials.gov: NCT05888493
. Updated 6 December 2024. Accessed 8 January 2025. https://clinicaltrials.gov/study/NCT05888493.
44.
A study to evaluate the efficacy and safety of liso-cel compared to standard of care in adults with relapsed or refractory follicular lymphoma. ClinicalTrials.gov identifier: NCT06313996
. Updated 15 March 2024. Accessed 8 January 2025. https://clinicaltrials.gov/study/NCT06313996.
45.
Nastoupil
LJ
,
Morschhauser
F
,
Scholz
CW
, et al
.
CELESTIMO: a phase III trial evaluating the efficacy and safety of mosunetuzumab plus lenalidomide versus rituximab plus lenalidomide in patients with relapsed or refractory follicular lymphoma who have received ≥ 1 line of systemic therapy [abstract]
.
J Clin Oncol
.
2022
;
40
(
suppl 16
):
TPS7588
.
46.
Falchi
L
,
Morschhauser
F
,
Linton
K
, et al
.
EPCORE FL-1: phase 3 trial of subcutaneous epcoritamab with rituximab and lenalidomide (R2) vs R2 alone in patients with relapsed or refractory follicular lymphoma [abstract]
.
Blood
.
2023
;
142
(
suppl 1
):
3053
.
47.
Vitolo
U
,
Phillips
EH
,
Alonso
AA
, et al
.
Trial in progress: phase 3 trial of odronextamab plus lenalidomide versus rituximab plus lenalidomide in relapsed/refractory follicular lymphoma and marginal zone lymphoma (olympia-5) [abstract]
.
Hemasphere
.
2023
;
7
(
S3
):
e49668c2
. Abstract PB2266.
48.
Salles
G
,
Shah
H
,
Leonard
JP
, et al
.
IBCL-485 a phase III, randomized, double-blind, study of tazemetostat or placebo plus lenalidomide and rituximab in patients with relapsed/refractory follicular lymphoma [abstract]
.
Clinical Lymphoma Myeloma and Leukemia
.
2023
;
23
(
suppl 1
):
S454
-
S455
.
49.
Zinzani
PL
,
Mayer
J
,
Flowers
CR
, et al
.
ROSEWOOD: a phase II randomized study of zanubrutinib plus obinutuzumab versus obinutuzumab monotherapy in patients with relapsed or refractory follicular lymphoma
.
J Clin Oncol
.
2023
;
41
(
33
):
5107
-
5117
.
50.
A study to compare the efficacy and safety of glocadomide in combination with rituximab (Golca+R) vs investigator’s choice in participants with relapsed/refractory follicular lymphoma (GOLSEEK-4). ClinicalTrials.gov identifier: NCT06911502
. Updated 7 May 2025. Accessed 17 May 2025. https://clinicaltrials.gov/study/NCT06425302.
51.
Neelapu
SS
,
Chavez
JC
,
Sehgal
A
, et al
.
Three-year follow-up analysis of axicabtagene ciloleucel in relapsed/refractory indolent non-Hodgkin lymphoma (ZUMA-5)
.
Blood
.
2024
;
143
(
6
):
496
-
506
.
52.
Dreyling
M
,
Fowler
NH
,
Dickinson
M
, et al
.
Durable response after tisagenlecleucel in adults with relapsed/refractory follicular lymphoma: ELARA trial update
.
Blood
.
2024
;
143
(
17
):
1713
-
1725
.
53.
Morschhauser
F
,
Dahiya
S
,
Palomba
ML
, et al
.
Lisocabtagene maraleucel in follicular lymphoma: the phase 2 TRANSCEND FL study
.
Nat Med
.
2024
;
30
(
8
):
2199
-
2207
.
54.
Sehn
LH
,
Luminari
S
,
Scholz
CW
, et al
.
Tafasitamab plus lenalidomide and rituximab for relapsed or refractory follicular lymphoma: results from a phase 3 study (inMIND) [abstract]
.
Blood
.
2024
;
144
(
suppl 2
):
LBA-1
.
55.
Morschhauser
F
,
Bishton
M
,
Eyre
TA
, et al
.
Mosunetuzumab in combination with lenalidomide has a manageable safety profile and encouraging activity in patients with relapsed/refractory follicular lymphoma: initial results from a phase Ib study [abstract]
.
Blood
.
2021
;
138
(
suppl 1
):
129
.
56.
Falchi
L
,
Sureda
A
,
Leppä
S
, et al
.
Fixed-duration epcoritamab + R2 drives deep and durable responses in patients with relapsed or refractory follicular lymphoma: 2-year follow-up from arm 2 of the Epcore NHL-2 trial [abstract]
.
Blood
.
2024
;
144
(
suppl 1
):
342
.
57.
Hou
J
,
Nair
R
,
Jacobs
R
, et al
.
Escalating doses of AZD0486, a novel CD19xCD3 T-cell engager, result in high complete remissions with rapid clearance of minimal residual disease in patients with relapsed/refractory follicular lymphoma [abstract]
.
Blood
.
2024
;
144
(
uppl 1
):
341
.
58.
Morschhauser
F
,
Tilly
H
,
Chaidos
A
, et al
.
Tazemetostat for patients with relapsed or refractory follicular lymphoma: an open-label, single-arm, multicentre, phase 2 trial
.
Lancet Oncol
.
2020
;
21
(
11
):
1433
-
1442
.
59.
Chavez
JC
,
Perini
GF
,
Nastoupil
L
, et al
.
Golcadomide (GOLCA) ± rituximab (RTX) demonstrates durable efficacy and is well tolerated in patients (pts) with relapsed/refractory follicular lymphoma (R/R FL): updated results from the phase 1/2 CC-99282-NHL-001 study [abstract]
.
Blood
.
2024
;
144
(
suppl 1
):
3018
.
60.
Thieblemont
C
,
Munoz
J
,
Tucci
A
, et al
.
Iberdomide (CC-220) monotherapy or in combination with an anti-CD20 monoclonal antibody as effective therapy in patients with relapsed/refractory lymphoma: early results from a phase 1/2 study [abstract]
.
Blood
.
2022
;
140
(
suppl 1
):
569
-
572
.
61.
Ferreri
CJ
,
Hildebrandt
MAT
,
Hashmi
H
, et al
.
Real-world experience of patients with multiple myeloma receiving ide-cel after a prior BCMA-targeted therapy
.
Blood Cancer J
.
2023
;
13
(
1
):
117
.
62.
Cohen
AD
,
Mateos
MV
,
Cohen
YC
, et al
.
Efficacy and safety of cilta-cel in patients with progressive multiple myeloma after exposure to other BCMA-targeting agents
.
Blood
.
2023
;
141
(
3
):
219
-
230
.
63.
Philipp
N
,
Kazerani
M
,
Nicholls
A
, et al
.
T-cell exhaustion induced by continuous bispecific molecule exposure is ameliorated by treatment-free intervals
.
Blood
.
2022
;
140
(
10
):
1104
-
1118
.
64.
Luminari
S
,
Manni
M
,
Galimberti
S
, et al;
Fondazione Italiana Linfomi
.
Response-adapted postinduction strategy in patients with advanced-stage follicular lymphoma: the FOLL12 study
.
J Clin Oncol
.
2022
;
40
(
7
):
729
-
739
.
65.
Kurtz
DM
,
Green
MR
,
Bratman
SV
, et al
.
Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing
.
Blood
.
2015
;
125
(
24
):
3679
-
3687
.
66.
Ching
T
,
Duncan
ME
,
Newman-Eerkes
T
, et al
.
Analytical evaluation of the clonoSEQ assay for establishing measurable (minimal) residual disease in acute lymphoblastic leukemia, chronic lymphocytic leukemia, and multiple myeloma
.
BMC Cancer
.
2020
;
20
(
1
):
612
.
67.
Roschewski
M
,
Dunleavy
K
,
Pittaluga
S
, et al
.
Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: a correlative biomarker study
.
Lancet Oncol
.
2015
;
16
(
5
):
541
-
549
.
68.
Kurtz
DM
,
Soo
J
,
Co Ting Keh
L
, et al
.
Enhanced detection of minimal residual disease by targeted sequencing of phased variants in circulating tumor DNA
.
Nat Biotechnol
.
2021
;
39
(
12
):
1537
-
1547
.
69.
Roschewski
M
,
Longo
DL
,
Armitage
JO
.
Circulating tumor DNA as measurable residual disease in aggressive B-cell lymphoma: a narrative review
.
JAMA Oncol
.
2025
;
11
(
4
):
416
-
422
.
70.
Roschewski
M
,
Kurtz
DM
,
Westin
J
, et al
.
MRD-negativity after frontline DLBCL therapy: pooled analysis of 6 clinical trials [abstract]
.
Hematol Oncol
.
2023
;
41
(
S2
):
177
-
179
.
71.
Roschewski
M
,
Lindenberg
L
,
Mena
E
, et al
.
End-of-treatment response assessment after frontline therapy for aggressive B-cell lymphoma: landmark comparison of a singular PET/CT scan versus ultrasensitive circulating tumor DNA [abstract]
.
Blood
.
2023
;
142
(
suppl 1
):
192
.
72.
Sworder
BJ
,
Yoon
SE
,
Kim
SJ
, et al
.
Prognostic utility of minimal residual disease (MRD) after curative intent induction therapy for DLBCL: a prospective real-world ctDNA study [abstract]
.
Blood
.
2023
;
142
(
suppl 1
):
69
.
73.
Lakhotia
R
,
Pittaluga
S
,
Phelan
JD
, et al
.
Circulating tumor DNA predicts time to first treatment in previously untreated follicular lymphoma: analysis from a prospective clonal evolution study [abstract]
.
Blood
.
2024
;
144
(
suppl 1
):
2958
.
74.
Fernández-Miranda
I
,
Pedrosa
L
,
Llanos
M
, et al
.
Monitoring of circulating tumor DNA predicts response to treatment and early progression in follicular lymphoma: results of a prospective pilot study
.
Clin Cancer Res
.
2023
;
29
(
1
):
209
-
220
.
75.
Park
JH
,
Geyer
MB
,
Brentjens
RJ
.
CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date
.
Blood
.
2016
;
127
(
26
):
3312
-
3320
.
76.
Bödör
C
,
Grossmann
V
,
Popov
N
, et al
.
EZH2 mutations are frequent and represent an early event in follicular lymphoma
.
Blood
.
2013
;
122
(
18
):
3165
-
3168
.
77.
Nastoupil
LJ
,
Song
Y
,
Sehn
LH
, et al
.
MAHOGANY: a phase 3 trial of zanubrutinib plus anti-CD20 antibodies vs lenalidomide plus rituximab in patients with relapsed or refractory follicular or marginal zone lymphoma [abstract]
.
J Clin Oncol
.
2023
;
41
(
suppl 16
):
TPS7590
.
78.
Chavez
JC
,
Nastoupil
LJ
,
Morschhauser
F
, et al
.
Efficacy and safety of golcadomide, a novel cereblon E3 ligase modulator (CELMoD) agent, combined with rituximab in a phase 1/2 open-label study of patients with relapsed/refractory non-Hodgkin lymphoma [abstract]
.
Blood
.
2023
;
142
(
suppl 1
):
4496
.
79.
Leonard
JP
,
Trneny
M
,
Izutsu
K
, et al
.
AUGMENT: a phase III study of lenalidomide plus rituximab versus placebo plus rituximab in relapsed or refractory indolent lymphoma
.
J Clin Oncol
.
2019
;
37
(
14
):
1188
-
1199
.
80.
Maurer
MJ
,
Bachy
E
,
Ghesquières
H
, et al
.
Early event status informs subsequent outcome in newly diagnosed follicular lymphoma
.
Am J Hematol
.
2016
;
91
(
11
):
1096
-
1101
.
81.
Westin
JR
,
Oluwole
OO
,
Kersten
MJ
, et al
.
Survival with axicabtagene ciloleucel in large B-cell lymphoma
.
N Engl J Med
.
2023
;
389
(
2
):
148
-
157
.
82.
Chavez
JC
,
Dickinson
M
,
Munoz
J
, et al
.
Three-year follow-up analysis of first-line axicabtagene ciloleucel for high-risk large B-cell lymphoma: ZUMA-12 study
.
Blood
.
2025
;
145
(
20
):
2303
-
2311
.
83.
Iacoboni
G
,
Navarro
V
,
Martín-López
, et al
.
Recent bendamustine treatment before apheresis has a negative impact on outcomes in patients with large B-cell lymphoma receiving chimeric antigen receptor T-cell therapy
.
J Clin Oncol
.
2024
;
42
(
2
):
205
-
217
.
84.
Iacoboni
G
,
Kyvsgaard
ER
,
Garces
VN
, et al
.
Impact of prior bendamustine exposure on bispecific antibody treatment outcomes in patients with relapsed/refractory follicular lymphoma [abstract]
.
Blood
.
2024
;
144
(
suppl 1
):
3009
.
85.
Iacoboni
G
,
Serna
A
,
Garces
VN
, et al
.
Impact of prior bendamustine exposure on bispecific antibody treatment outcomes for patients with B-cell lymphoma [abstract]
.
Blood
.
2023
;
142
(
suppl 1
):
310
.
86.
Tobin
JW
,
Chikatamarla
VA
,
Matic
M
, et al
.
Outcomes for high-risk defining events in follicular lymphoma following frontline immunochemotherapy
.
Blood Neoplasia
.
2024
;
1
(
4
):
100044
.
87.
Akhtar
OS
,
Huang
LW
,
Tsang
M
, et al
.
A Young International Society of Geriatric Oncology (YSIOG) review paper
.
J Geriatr Oncol
.
2022
;
13
(
5
):
572
-
581
.
88.
Tsang
M
,
Noble
BN
,
Bommier
C
, et al
.
Frontline management strategy and quality of life in follicular lymphoma: a multi-institutional prospective cohort study [abstract]
.
Blood
.
2023
;
142
(
suppl 1
):
3761
.
89.
Tsang
M
,
Durani
U
,
Day
JR
, et al
.
Understanding the patient perspective on treatment preferences for follicular lymphoma [abstract]
.
Blood
.
2024
;
144
(
suppl 1
):
7598
.
90.
Thomas
C
,
Marsh
K
,
Trapali
M
, et al
.
Preferences of patients and physicians in the United States for relapsed/refractory follicular lymphoma treatments
.
Cancer Med
.
2024
;
13
(
19
):
e70177
.
91.
Gaballa
S
,
Xue
M
,
Totev
TI
, et al
.
Patient medication preferences in follicular lymphoma (FL) in the United States (USA): a discrete-choice experiment (DCE) [abstract]
.
Blood
.
2024
;
144
(
suppl
):
3655
.
92.
Johnson
PC
,
Bailey
A
,
Ma
Q
, et al
.
Quality of life evaluation in patients with follicular cell lymphoma: a real-world study in Europe and the United States
.
Adv Ther
.
2024
;
41
(
8
):
3342
-
3361
.

Author notes

Data are available on request from the corresponding author, Carla Casulo (carla_casulo@urmc.rochester.edu).