Patients with relapsed and refractory (R/R) aggressive B-cell non-Hodgkin lymphomas have historically poor survival outcomes, with chimeric antigen receptor T-cell (CAR-T) therapy now presenting a curative option for a subset of those patients. However, with the approval of several novel bispecific monoclonal antibody (BsAb) therapies with considerable activity in R/R aggressive large B-cell lymphomas (LBCL), patients and oncologists will be faced with decisions regarding how to sequence CAR-T and BsAb therapies based on patient- and disease-related factors. In this review, we compare CAR-T and BsAb therapies for R/R LBCL, highlighting data on the efficacy and toxicity of each treatment paradigm, and provide a roadmap for sequencing these highly effective therapies.

Learning Objectives

  • Understand the efficacy and safety of chimeric antigen receptor T-cell (CAR-T) and bispecific antibody therapies for aggressive large B-cell lymphoma in the third-line setting

  • Examine the real-world efficacy and safety of CAR-T therapies in patients who were not eligible for the initial registrational trials

  • Compare advantages and disadvantages of initial sequencing of CAR-T first vs bispecific antibodies first in relapsed and refractory large B-cell lymphomas

Survival of patients with relapsed and refractory (R/R) aggressive large B-cell lymphomas (LBCLs) was historically dismal in the chemotherapy era. However, the regulatory approval of chimeric antigen receptor T-cell (CAR-T) therapy has not only improved outcomes for heavily pretreated patients with R/R LBCL but also heralded a new epoch of immunotherapeutic trials in non-Hodgkin lymphomas (NHLs). Leading these trials are CD20-directed bispecific monoclonal antibody (BsAb) therapies, which have shown excellent efficacy and safety in R/R LBCL. With new regulatory approval of several BsAb agents, it is imperative to conceptualize a roadmap for sequencing these agents with CAR-T therapy in the treatment paradigm of aggressive LBCL. In this review, we will compare CAR-T and BsAb therapies for R/R LBCL and propose a treatment algorithm to guide practicing clinicians, highlighting data and arguments for the use of CAR-T first vs BsAb first in R/R LBCL.

A 70-year-old man with a good performance status and history of atrial fibrillation was diagnosed with stage IV diffuse large B-cell lymphoma (DLBCL) (germinal center cell of origin, MYC-amplified) 3 years ago and received 6 cycles of rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone with a complete response (CR) at the end of treatment. He relapsed 15 months later and subsequently received second-line chemotherapy followed by autologous stem cell transplantation (ASCT). He did well for 11 months when he unfortunately developed a biopsy-proven relapse of DLBCL.

There are 3 CAR-T constructs that are approved by the US Food and Drug Administration (FDA) in the third-line setting for LBCL: axicabtagene ciloleucel (axi-cel) in 2017, tisagenlecleucel (tisa-cel) in 2018, and lisocabtagene maraleucel (liso-cel) in 2021. These FDA approvals were based on the ZUMA-1, JULIET, and TRANSCEND trials (Table 1). The long-term curative potential for CAR-T therapy has been demonstrated with axi-cel, with 5-year follow-up data demonstrating a progression-free survival (PFS) plateau of 32% and a disease-specific survival of 51%,1  as well as with CTL019 (tisa-cel) with a 5-year PFS of 31%.2  Although clearly with curative potential, CAR-T therapy can be associated with unique toxicities, with cytokine release syndrome (CRS) and neurotoxicity noted with all CAR-T constructs, as well as prolonged cytopenias in up to 45% of cases which can impact post–CAR-T outcomes.3  The TRANSCEND trial included more broad inclusion criteria than the other trials, including patients with older age, secondary central nervous system (CNS) involvement, and mild renal and cardiac insufficiency, and with no minimum absolute lymphocyte count requirement for apheresis. The efficacy and toxicity of CAR-T products in the third-line setting have not been directly compared. Indirect comparisons between the ZUMA-1 and TRANSCEND trials have demonstrated comparable efficacy between axi-cel and liso-cel with lower rates of toxicities with liso-cel, underscoring differing functions of the constructs' costimulatory domains (CD28 vs 4-1BB).4 

Table 1.

Clinical trials of CAR T-cell and bispecific antibodies therapies in the third-line setting for aggressive LBCL

Clinical trialConstructPatientsHistologiesResponse ratesSurvival outcomesLongest median follow-upDuration of responseRates of CRSRates of neurotoxicityTreatment-related mortalityFDA approved
CAR-T therapies for third-line LBCL 
ZUMA-11,5  Axi-cel 101 DLBCL, PMBCL, tFL ORR 82% (CR 54%) mPFS 5.9 mo
12 mo:
PFS 44%, OS 59%
5 y:
PFS 32%, OS 43% 
63.1 mo mDOR 11.1 mo
mDOCR 62.2 mo 
93%
(13% grade 3+) 
64%
(28% grade 3+) 
3 patients (3%), 1 death due to CRS, 1 death due to HLH Yes 
JULIET2,6  tisa-cel 115 DLBCL, tFL, HGBL ORR 53% (CR 39%) mPFS 2.9 mo
12 mo:
RFS 65%,
OS 49%
40 mo:
PFS 38%,
OS 39%
5 y:
PFS 31% 
60.7 mo mDOR NR 58%
(22% grade 3+) 
21%
(12% grade 3+) 
0 patients Yes 
TRANSCEND7  Liso-cel 256 DLBCL, tFL, HGBL, PMBCL, FL 3B ORR 73% (CR 53%) mPFS 6.8 mo
1 y:
PFS 44%,
OS 58%
 
18.8 mo mDOR 17 mo 42%
(2% grade 3+) 
30%
(10% grade 3+) 
7 patients (3%) Yes 
Bispecific antibody therapies for third-line LBCL 
NCT030756968,9  Glofitamab 154
51 (33%) with prior CAR-T exposure 
DLBCL, tFL, HGBL, PMBCL ORR 52% (CR 39%)
Prior CAR-T: CR 35% 
mPFS 4.9 mo
1 y:
PFS 37%,
OS 50%
 
12.6 mo mDOR 18.4 mo
mDOCR NR (74% remained in CR at median of 18.1 mo of follow-up) 
63% (4% grade 3+) 8% (3% grade 3+) 8 patients (5%) Yes (June 2023) for LBCL after at least 2 lines of therapy 
EPCORE NHL-1 (NCT03625037)10,11  Epcoritamab (dose escalation) 68
46 with LBCL 
Any relapsed or refractory CD20+ mature B-cell NHL ORR 68% (CR 45%) for LBCL mPFS for LBCL: 9.1 mo 9.3 mo 75% of LBCL responders in ongoing remission at 6 mo 59% (no grade 3+) 6% (3% grade 3+) 0 patients Yes (May 2023) for LBCL after at least 2 lines of therapy 
 Epcoritamab (dose expansion) 157
61 (39%) with prior CAR-T exposure 
DLBCL, PMBCL, HGBL, FL 3B ORR 63% (CR 39%)
Prior CAR-T:
ORR 54% (CR 34%) 
mPFS 4.4 mo
6 mo:
PFS 44%
mPFS NR among patients with CR 
10.7 mo mDOR
12 mo (9.7 mo if prior CAR-T)
mDOCR NR (NR if prior CAR-T) 
50% (2.5% grade 3+) 6% (0.6% grade 3+) 9 patients (6%), 1 death due to ICANS  
NCT0250040712,13  Mosunetuzumab (phase I data) 197
129 with aggressive NHL 
Any relapsed or refractory B-cell NHL ORR 35% (CR 19%) mPFS 1.4 mo 11.9 mo mDOR
7.6 mo
mDOCR
23 mo
 
27% (1% grade 3+) 10-18% (4% grade 3+) 3 patients (1.5%) Yes, for relapsed or refractory FL 
 Mosunetuzumab (phase II data) 88 DLBCL, tFL, HGBL ORR 42% (CR 24%)
Prior CAR-T:
ORR 23% (CR 12%) 
mPFS 3.2 mo
mOS 11.5 mo
Prior CAR-T:
mPFS 1.4 mo 
10.1 mo mDOR 7.0 mo
mDOCR NR 
26% (2.3% grade 3+) 1% (no grade 3+) 3 patients (3.4%)  
ELM-1
(NCT02290951)14  
Odronextamab 85 with LBCL
33 (39%) of LBCL with prior CAR-T exposure 
Any relapsed or refractory B-cell NHL No prior CAR-T:
ORR 53% (CR 53%)
Prior CAR-T:
ORR 33% (CR 27%) 
No prior CAR-T:
mPFS 11.5 mo
Prior CAR-T:
mPFS 2.0 mo 
4.2 mo No prior CAR-T:
mDOR NR
Prior CAR-T:
mDOR NR 
54% (7% grade 3+) 12% (3% grade 3+) 7 patients (5%) No 
Clinical trialConstructPatientsHistologiesResponse ratesSurvival outcomesLongest median follow-upDuration of responseRates of CRSRates of neurotoxicityTreatment-related mortalityFDA approved
CAR-T therapies for third-line LBCL 
ZUMA-11,5  Axi-cel 101 DLBCL, PMBCL, tFL ORR 82% (CR 54%) mPFS 5.9 mo
12 mo:
PFS 44%, OS 59%
5 y:
PFS 32%, OS 43% 
63.1 mo mDOR 11.1 mo
mDOCR 62.2 mo 
93%
(13% grade 3+) 
64%
(28% grade 3+) 
3 patients (3%), 1 death due to CRS, 1 death due to HLH Yes 
JULIET2,6  tisa-cel 115 DLBCL, tFL, HGBL ORR 53% (CR 39%) mPFS 2.9 mo
12 mo:
RFS 65%,
OS 49%
40 mo:
PFS 38%,
OS 39%
5 y:
PFS 31% 
60.7 mo mDOR NR 58%
(22% grade 3+) 
21%
(12% grade 3+) 
0 patients Yes 
TRANSCEND7  Liso-cel 256 DLBCL, tFL, HGBL, PMBCL, FL 3B ORR 73% (CR 53%) mPFS 6.8 mo
1 y:
PFS 44%,
OS 58%
 
18.8 mo mDOR 17 mo 42%
(2% grade 3+) 
30%
(10% grade 3+) 
7 patients (3%) Yes 
Bispecific antibody therapies for third-line LBCL 
NCT030756968,9  Glofitamab 154
51 (33%) with prior CAR-T exposure 
DLBCL, tFL, HGBL, PMBCL ORR 52% (CR 39%)
Prior CAR-T: CR 35% 
mPFS 4.9 mo
1 y:
PFS 37%,
OS 50%
 
12.6 mo mDOR 18.4 mo
mDOCR NR (74% remained in CR at median of 18.1 mo of follow-up) 
63% (4% grade 3+) 8% (3% grade 3+) 8 patients (5%) Yes (June 2023) for LBCL after at least 2 lines of therapy 
EPCORE NHL-1 (NCT03625037)10,11  Epcoritamab (dose escalation) 68
46 with LBCL 
Any relapsed or refractory CD20+ mature B-cell NHL ORR 68% (CR 45%) for LBCL mPFS for LBCL: 9.1 mo 9.3 mo 75% of LBCL responders in ongoing remission at 6 mo 59% (no grade 3+) 6% (3% grade 3+) 0 patients Yes (May 2023) for LBCL after at least 2 lines of therapy 
 Epcoritamab (dose expansion) 157
61 (39%) with prior CAR-T exposure 
DLBCL, PMBCL, HGBL, FL 3B ORR 63% (CR 39%)
Prior CAR-T:
ORR 54% (CR 34%) 
mPFS 4.4 mo
6 mo:
PFS 44%
mPFS NR among patients with CR 
10.7 mo mDOR
12 mo (9.7 mo if prior CAR-T)
mDOCR NR (NR if prior CAR-T) 
50% (2.5% grade 3+) 6% (0.6% grade 3+) 9 patients (6%), 1 death due to ICANS  
NCT0250040712,13  Mosunetuzumab (phase I data) 197
129 with aggressive NHL 
Any relapsed or refractory B-cell NHL ORR 35% (CR 19%) mPFS 1.4 mo 11.9 mo mDOR
7.6 mo
mDOCR
23 mo
 
27% (1% grade 3+) 10-18% (4% grade 3+) 3 patients (1.5%) Yes, for relapsed or refractory FL 
 Mosunetuzumab (phase II data) 88 DLBCL, tFL, HGBL ORR 42% (CR 24%)
Prior CAR-T:
ORR 23% (CR 12%) 
mPFS 3.2 mo
mOS 11.5 mo
Prior CAR-T:
mPFS 1.4 mo 
10.1 mo mDOR 7.0 mo
mDOCR NR 
26% (2.3% grade 3+) 1% (no grade 3+) 3 patients (3.4%)  
ELM-1
(NCT02290951)14  
Odronextamab 85 with LBCL
33 (39%) of LBCL with prior CAR-T exposure 
Any relapsed or refractory B-cell NHL No prior CAR-T:
ORR 53% (CR 53%)
Prior CAR-T:
ORR 33% (CR 27%) 
No prior CAR-T:
mPFS 11.5 mo
Prior CAR-T:
mPFS 2.0 mo 
4.2 mo No prior CAR-T:
mDOR NR
Prior CAR-T:
mDOR NR 
54% (7% grade 3+) 12% (3% grade 3+) 7 patients (5%) No 

FL 3B, grade 3B follicular lymphoma; HGBL, high-grade B-cell lymphoma; HLH, hemophagocytic lymphohistiocystosis; ICANS, immune effector cell-associated neurotoxicity syndrome; mDOCR, median duration of complete response; mDOR, median duration of response; mOS, median overall survival; mPFS, median progression-free survival; NR, not reached; PMBCL, primary mediastinal large B-cell lymphoma; tFL, transformed follicular lymphoma.

ASCT is curative in less than 30% of patients with transplant-eligible R/R LBCL in the second-line setting, and its efficacy is particularly poor for patients with primary refractory or early-relapsed LBCL.15  Given the considerable activity of CAR-T therapy in LBCL in the third-line setting, especially for patients with relapse after ASCT, it was intuitive to assess the efficacy of CAR-T in the second- line setting in patients with high-risk R/R LBCL. There have been 3 large randomized controlled trials of CAR-T vs standard of care (SOC) in the second-line setting, with SOC consisting of second- line chemoimmunotherapy followed by ASCT in transplant- eligible high-risk primary refractory or early-relapsed LBCL within 12 months of initial therapy: ZUMA-7, BELINDA, and TRANFORM. ZUMA-7 and TRANSFORM met their primary end points of event-free survival, while BELINDA did not. In ZUMA-7, 359 patients with high-risk R/R LBCL were randomized to axi-cel vs SOC.16  Median event-free survival (EFS) was significantly higher with axi-cel at 8.3 months compared to 2 months for standard care, with 2-year EFS of 41% and 16% for axi-cel and standard care, respectively. Improved outcomes with axi-cel were also observed in patients aged ≥65 years.17  Although there was no significant difference in overall survival (OS) initially reported, updated statistically significant OS results have now been reported in favor of axi-cel.18  The TRANSFORM trial of 184 patients randomized to liso-cel vs SOC utilized a similar trial design as ZUMA-7, although crossover to the liso-cel arm was allowed per protocol. Similarly, median EFS was superior with liso-cel (not reached vs 2.4 months) with 18-month EFS of 53% and 21% for liso-cel and standard care, respectively.19  In a prespecified OS analysis adjusting for crossover, there was a trend toward improvement in OS in the liso-cel arm, with 18-month OS rates of 73% for liso-cel and 54% for SOC. In both ZUMA-7 and TRANSFORM, there were no deaths due to CRS or neurotoxicity.

Given the favorable efficacy and toxicity profile of liso-cel in transplant-eligible R/R LBCL, PILOT was a phase 2 single-arm trial that investigated the efficacy of liso-cel in 74 transplant- ineligible patients with R/R LBCL in the second-line setting.20  The overall response rate (ORR) was 80% (CR 54%) with a median PFS and OS of 9 months and not reached, respectively; for patients who achieved a CR, median PFS and OS were 23 months and not reached, respectively.

Based on the positive results of the ZUMA-7, TRANSFORM, and PILOT trials, the FDA approved both axi-cel and liso-cel for patients with high-risk LBCL in first relapse, as well as liso-cel in transplant-ineligible patients after failure of 1 line of therapy.

BsAb therapies cotarget tumor antigens and endogenous immune effector cells to induce tumor killing by recruiting and activating peripheral and intratumoral T cells, natural killer cells, and/or macrophages to the tumor microenvironment. In B-cell lymphomas, several off-the-shelf IgG-type BsAb agents have been developed that cotarget CD20 and CD3 for T-cell–mediated cytotoxicity—namely, mosunetuzumab, glofitamab, epcoritamab, and odronextamab (Table 1). Both glofitamab and epcoritamab have been studied in third-line LBCL and received FDA approval in mid-2023. In a phase 2 study of 154 patients with LBCL who received up to 12 cycles of fixed-duration intravenous glofitamab after at least 2 prior lines of therapy, ORR was 52% (CR 39%) with 1-year PFS and OS of 37% and 50%, respectively.8  Importantly, the 33% of patients in the study who had previously received CAR-T had a similar CR rate of 35%. Most patients who achieved a CR had durable remissions, with a median duration of CR not reached at 18.1 months of follow-up.9  Although CRS was common (63%), only 4% were high-grade events and none were grade 5; any-grade neurotoxicity occurred in 8% of patients with only 3% of high grade.

In a phase 1/2 study of 157 patients with LBCL after at least 2 lines of previous therapy who received subcutaneous epcoritamab until disease progression or unacceptable toxicity, ORR was 63% (CR 39%) with a median duration of response (DOR) of 12 months and 12-month PFS of 38%.10,11  Thirty-nine percent of patients had previously received CAR-T with an ORR of 54% (CR 34%) with median DOR of 9.7 months. Similar to glofitamab, CRs were durable after epcoritamab with median duration of CR not reached at 10.7 months of follow-up, including in post–CAR-T patients. CRS was also common with epcoritamab (50%), and although only 2.5% of events were high-grade CRS with no grade 5 events, there was 1 treatment-related death due to neurotoxicity in the study.

Although mosunetuzumab was FDA approved in December 2022 for R/R follicular lymphoma, activity was significantly lower in LBCL in a phase 1 study, with ORR 35% (CR 19%) as compared to ORR 66% (CR 49%) in indolent NHL, with a median PFS of 1.4 months in aggressive NHL.12  Follow-up phase 2 data from this study in 88 patients with R/R LBCL demonstrated ORR 42% (CR 24%) with a median PFS of 3.2 months, with a lower CR rate of 12% in patients who had previously received CAR-T.13  Combinations of other agents with mosunetuzumab have been investigated to improve efficacy, including polatuzumab vedotin with mosunetuzumab in R/R LBCL, which has demonstrated an ORR of 72% (CR 56%) in older patients who may not be candidates for CAR-T or who are relapsed after CAR-T.21 

The durable remissions achieved in patients with heavily pretreated and poor-prognosis LBCL, including after CAR-T and with fixed-duration regimens such as glofitamab, have generated considerable interest in BsAb therapies, particularly given the low incidence of high-grade CRS and neurotoxicity. Further, both glofitamab and epcoritamab demonstrated a relatively fast time to response of 1.4 months, which is particularly important for an off-the-shelf therapeutic that does not require manufacturing. Other novel CD20xCD3 BsAb agents are under development, including odronextamab and plamotamab with ORR 33% to 53% (CR 27%-53%) and ORR 47% (CR 26%) in LBCL, respectively.14,22  Given the encouraging results of these therapies in heavily pretreated LBCL, several trials with BsAb therapies are ongoing in earlier lines of therapy either as single agents or in combination with chemotherapy. These results are awaited to clarify how BsAb therapies may be best sequenced after first-line therapy.23 

The primary advantage of sequencing CAR-T therapy before BsAb therapy in the third-line LBCL setting is the potential for cure with CAR-T in approximately one-third of patients at 5 years of follow-up, which has not yet been demonstrated with BsAb agents. However, given the higher risk of toxicities with CAR-T compared to BsAb therapies, particularly CRS and neurotoxicity (Table 1) as well as prolonged cytopenias and infections, there is concern that administering CAR-T to patients who did not meet the inclusion criteria of the initial trials may incur excess toxicity and mortality. Several retrospective, “real-world” studies of CAR-T outcomes have demonstrated comparable survival and safety outcomes when CAR-T was administered to patients who would have been ineligible for the original clinical trials due to advanced age, comorbidities, or performance status (Table 2). These real-world studies also demonstrated durable remissions in patients who achieved CR, suggesting the possibility for cure in trial-ineligible patients.24,25  Toxicities also appear to be comparable outside of the trial setting, with preserved survival rates despite higher use of tocilizumab and steroids in the real-world setting.24  Longitudinal improvements in quality of life and patient-reported outcomes after CAR-T have been demonstrated in both clinical trials and real-world studies,26,27  further supporting the tolerability of CAR-T outside of the trial setting.

Table 2.

Real-world CAR-T outcomes in third-line LBCL

Real-world study designConstructsPatientsPercent of patients ineligible for CAR-T clinical trialsResponse ratesSurvival outcomesMedian duration of responseRates of CRSRates of neurotoxicityTreatment-related mortalityConclusions
Locke et al.28 
Postapproval safety observational study 
Axi-cel 1343 38% aged ≥65, 4% with ECOG PS 2+, 13% cardiac comorbidities, 2% hepatic comorbidities, 2% renal comorbidities, 15% double/ triple-hit lymphoma, 66% refractory disease ORR 74% (CR 56%)
ORR 78% (CR 62%) for patients aged ≥65
ORR 57% (CR 29%) for hepatic comorbidities
ORR 70% (CR 43%) for renal comorbidities
ORR 47% (CR 20%) for ECOG 2-3 
18 mo:
PFS 42%, OS 52% 
18 mo:
DOR 61% 
83% 55% Not assessed Patients aged ≥65 with moderate to severe pulmonary disease and ECOG 2-3 had inferior ORR.
Age ≥65 was not associated with inferior survival, but was associated with higher rates of CRS and ICANS.
ECOG PS significantly affected all efficacy outcomes. 
Jacobson et al.25 
Postauthorization safety study through the CIBMTR registry 
Axi-cel 1297 57% were ineligible for ZUMA-1 ORR 73% (CR 56%) mPFS 8.6 mo
mOS 21.8 mo 
mDOR NR
24-mo DOR 57% 
83% (8% grade 3+) 55% (24% grade 3+) 3%, 2% died from CRS, 1% died from ICANS Real-world response rates were similar to ZUMA-1, with similar DOR in patients who were and were not eligible for ZUMA-1.
High-grade CRS and ICANS were lower in the real-world cohort.
ECOG PS of 2 or greater was associated with inferior response rates, PFS, and OS.
Patients age ≥65 had a higher ORR despite higher risk of CRS and ICANS as compared to younger patients. 
Landsburg et al.29 
Observational study through the CIBMTR registry 
Tisa-cel 1159 31% ineligible for JULIET ORR 60% 2 y:
PFS 28%, OS 44% 
2 y:
DOR 53% 
58% (6% grade 3+) 23% (7% grade 3+) Not assessed Real-world outcomes were similar to JULIET.
Patients with comorbidities (who were not eligible for JULIET) had similar efficacy outcomes.
Patients with ECOG PS 2-4 had higher rates of high-grade CRS and ICANS but similar efficacy outcomes. 
Bachy et al.30 
Retrospective French DESCAR-T registry study with propensity score matching between axi-cel and tisa-cel 
Axi-cel
Tisa-cel 
452 axi-cel
277 tisa-cel 
Not assessed Axi-cel:
ORR 80% (CR 60%)
Tisa-cel:
ORR 66% (CR 42%) 
1 y:
PFS 47% axi-cel, 33% tisa-cel
OS 64% axi-cel, 49% tisa-cel
 
1 y:
DOR 54% axi-cel, 42% tisa-cel 
Axi-cel:
86% (5% grade 3+)
Tisa-cel:
76% (9% grade 3+) 
Axi-cel:
49% (14% grade 3+)
Tisa-cel:
22% (3% grade 3+) 
Axi-cel: no grade 5 CRS, 1 patient grade 5 ICANS
Tisa-cel: 2 grade 5 CRS, no grade 5 ICANS
No other treatment-related grade 5 AEs 
Real-world response and survival rates were similar to clinical trials.
Real-world ORR, CR, PFS, and OS better with axi-cel compared to tisa-cel, although axi-cel with more toxicity. 
Chihara et al.31 
Retrospective cohort from Medicare claims data 
All CAR-T 551 31% of patients in this cohort were age ≥75 Not assessed Age ≥75:
mPFS 160 d
mOS 403 d
Age 70-74:
mPFS 379 d
mOS 603 d
Age 65-69:
mPFS 194
mOS 518
1 y PFS:
Age ≥75 34%
Age 70-74 52%
Age 65-69 43% 
Not assessed Not assessed Not assessed Not assessed Older patients, particularly those age ≥75, had significantly worse PFS and OS compared to younger patients as well as compared to clinical trials. 
Nastoupil et al.32 
Retrospective cohort from the US Lymphoma CAR-T Consortium 
Axi-cel 298 43% were ineligible for ZUMA-1 due to comorbidities ORR 82% (CR 64%) 1 y:
PFS 47%
OS 68%
PFS 34% for ZUMA-1 ineligible patients 
mDOR NR (median follow-up of 12.9 mo) 91% (7% grade 3+) 69% (31% grade 3+ 4.4%, 1 death due to HLH, 1 death due to ICANS Real-world outcomes and safety were comparable to ZUMA-1, although ECOG PS 2-4 had worse PFS, OS, and toxicities.
Patients ineligible for ZUMA-1 had inferior PFS and OS. 
Sano et al.33 
Retrospective cohort from the US Lymphoma CAR-T Consortium 
Axi-cel 272
30% were aged ≥65 
Not assessed Age ≥65
ORR 84% (CR 71%)
Age ≤65
ORR 82% (CR 51%) 
Age ≥65
mPFS 9.2 mo, mOS not assessable
Age ≤65
mPFS 7.4 mo, mOS 18.7 mo 
Not assessed Age ≥65
92% (7% grade 3+)
Age ≤65
91% (7% grade 3+) 
Age ≥65
78% (35% grade 3+)
Age ≤65
65% (31% grade 3+) 
2 deaths (1 in age ≥65 and 1 age <65) Rate of complete response was higher in patients aged ≥65 compared to age <65.
There was no difference in PFS, OS, and toxicities between patients aged <65 and ≥65. 
Bethge et al.34 
Retrospective cohort from the German Registry for Stem Cell Transplantation 
Axi-cel
Tisa-cel 
173 axi-cel
183 tisa-cel 
13% eligible for ZUMA-1
89% eligible for JULIET 
Axi-cel:
ORR 74% (CR 42%)
Tisa-cel:
ORR 53% (CR 32%) 
Axi-cel:
1 y PFS 35%, OS 55%
Tisa-cel:
1 y PFS 24%, OS 53% 
Not assessed Axi-cel:
81% (10% grade 3+)
Tisa-cel:
65% (13% grade 3+) 
Axi-cel:
44% (16% grade 3+)
Tisa-cel:
22% (7% grade 3+) 
Axi-cel:
2 y
10.4%
Tisa-cel:
2 y 3.5% 
Real-world ORR, CR, and OS were comparable to clinical trials, but real-world PFS was worse.
Higher rates of delayed infection-related NRM in real-world cohort. 
Riedell et al.35 
Retrospective cohort of 8 US centers
 
Axi-cel
Tisa-cel 
168 axi-cel
92 tisa-cel 
61% axi-cel ineligible for ZUMA-1
43% tisa-cel ineligible for JULIET 
Axi-cel:
ORR 52% (CR 44%)
Tisa-cel:
ORR 41% (CR 35%)
 
Axi-cel:
1 y PFS 42%, OS 62%
Tisa-cel:
1 y PFS 32%, OS 59% 
Axi-cel:
1 y DOR 70%
Tisa-cel:
1 y DOR 75% 
Axi-cel:
85% (9% grade 3+)
Tisa-cel:
39% (1% grade 3+)
mDOR NR for either cohort 
Axi-cel:
56% (38% grade 3+)
Tisa-cel:
11% (1% grade 3+) 
Axi-cel:
9%, 4 deaths from ICANS
Tisa-cel:
7%
 
Real-world outcomes were slightly lower compared to ZUMA-1 and JULIET, although safety outcomes were comparable.
Axi-cel and tisa-cel had comparable efficacy, although less CRS and ICANS with tisa-cel. 
Pasquini et al.36 
Postauthorization safety study through the CIBMTR registry 
Tisa-cel 155 Not assessed ORR 62% (CR 40%) 1 y:
EFS 52%
OS 77% 
1 y DOR 61% 45% (4.5% grade 3+) 18% (5.1% grade 3+) 1.2% Similar efficacy and improved safety compared to JULIET. 
Kittai et al.37 
Retrospective cohort from 4 centers 
Axi-cel
Tisa-cel 
94 axi-cel
36 tisa-cel 
57% of patients had high comorbidity based on CIRS score ORR 68% (CR 42%) mPFS 6.7 mo
mOS NR
1 y OS 60% 
Not assessed 79% 58% 12.3%, 2 deaths due to CRS, 1 death due to ICANS Worse ECOG PS and comorbidities by CIRS score were associated with worse survival.
Presence of more comorbidities was not associated with worse CRS or ICANS. 
Cook et al.38 
Meta-analysis of prospective and retrospective studies of CAR-T in primary and secondary CNS lymphoma 
Multiple CAR-T products 30 patients with primary CNS lymphoma (PCNSL)
98 patients with secondary CNS lymphoma (SCNSL) 
Not assessed PCNSL:
ORR 64% (CR 56%)
SCNSL:
ORR 57% (CR 47%) 
PCNSL:
6-mo PFS 37%
SCNSL:
6-mo PFS 37% 
PCNSL:
mDOR 9 mo
SCNSL:
mDOR 4.6 mo 
PCNSL:
70% (13% grade 3+)
SCNSL:
72% (11% grade 3+) 
PCNSL:
53% (18% grade 3+)
SCNSL:
48% (26% grade 3+) 
Not assessed Incidence of CRS and ICANS in real-world cohort of PCNSL and SCNSL is comparable to clinical trials.
Approximately one-third of patients with PCNSL and SCNSL had 6-mo complete responses, which could be durable with longer follow-up. 
Lin et al.50 
Single-center retrospective cohort of older adults who did and did not receive CAR-T 
Axi-cel and tisa-cel 24 older adults received CAR-T
18 older adults did not receive CAR-T
25 younger adults (age <65) received CAR-T 
Not assessed 100-d CR 51% (for 49 patients who received CAR-T) 6-mo PFS 48%, OS 71% (for 49 patients who received CAR-T)
Older adults who received CAR-T had a significantly lower risk of death (HR 0.31) compared to older adults who did not receive CAR-T
No difference in PFS or OS between older or younger adults who received CAR-T by chronological age, functional limitations, cognitive impairment, or comorbidity burden 
Not assessed 83% (8% grade 2±) 54% (25% grade 2±) 2 deaths in older adults after CAR-T Older adults who receive CAR-T have better postrelapse OS compared to patients who instead receive chemotherapy and/or supportive care.
PFS, OS, and rates of CRS and ICANS are not different between older and younger adults who receive CAR-T. 
Real-world study designConstructsPatientsPercent of patients ineligible for CAR-T clinical trialsResponse ratesSurvival outcomesMedian duration of responseRates of CRSRates of neurotoxicityTreatment-related mortalityConclusions
Locke et al.28 
Postapproval safety observational study 
Axi-cel 1343 38% aged ≥65, 4% with ECOG PS 2+, 13% cardiac comorbidities, 2% hepatic comorbidities, 2% renal comorbidities, 15% double/ triple-hit lymphoma, 66% refractory disease ORR 74% (CR 56%)
ORR 78% (CR 62%) for patients aged ≥65
ORR 57% (CR 29%) for hepatic comorbidities
ORR 70% (CR 43%) for renal comorbidities
ORR 47% (CR 20%) for ECOG 2-3 
18 mo:
PFS 42%, OS 52% 
18 mo:
DOR 61% 
83% 55% Not assessed Patients aged ≥65 with moderate to severe pulmonary disease and ECOG 2-3 had inferior ORR.
Age ≥65 was not associated with inferior survival, but was associated with higher rates of CRS and ICANS.
ECOG PS significantly affected all efficacy outcomes. 
Jacobson et al.25 
Postauthorization safety study through the CIBMTR registry 
Axi-cel 1297 57% were ineligible for ZUMA-1 ORR 73% (CR 56%) mPFS 8.6 mo
mOS 21.8 mo 
mDOR NR
24-mo DOR 57% 
83% (8% grade 3+) 55% (24% grade 3+) 3%, 2% died from CRS, 1% died from ICANS Real-world response rates were similar to ZUMA-1, with similar DOR in patients who were and were not eligible for ZUMA-1.
High-grade CRS and ICANS were lower in the real-world cohort.
ECOG PS of 2 or greater was associated with inferior response rates, PFS, and OS.
Patients age ≥65 had a higher ORR despite higher risk of CRS and ICANS as compared to younger patients. 
Landsburg et al.29 
Observational study through the CIBMTR registry 
Tisa-cel 1159 31% ineligible for JULIET ORR 60% 2 y:
PFS 28%, OS 44% 
2 y:
DOR 53% 
58% (6% grade 3+) 23% (7% grade 3+) Not assessed Real-world outcomes were similar to JULIET.
Patients with comorbidities (who were not eligible for JULIET) had similar efficacy outcomes.
Patients with ECOG PS 2-4 had higher rates of high-grade CRS and ICANS but similar efficacy outcomes. 
Bachy et al.30 
Retrospective French DESCAR-T registry study with propensity score matching between axi-cel and tisa-cel 
Axi-cel
Tisa-cel 
452 axi-cel
277 tisa-cel 
Not assessed Axi-cel:
ORR 80% (CR 60%)
Tisa-cel:
ORR 66% (CR 42%) 
1 y:
PFS 47% axi-cel, 33% tisa-cel
OS 64% axi-cel, 49% tisa-cel
 
1 y:
DOR 54% axi-cel, 42% tisa-cel 
Axi-cel:
86% (5% grade 3+)
Tisa-cel:
76% (9% grade 3+) 
Axi-cel:
49% (14% grade 3+)
Tisa-cel:
22% (3% grade 3+) 
Axi-cel: no grade 5 CRS, 1 patient grade 5 ICANS
Tisa-cel: 2 grade 5 CRS, no grade 5 ICANS
No other treatment-related grade 5 AEs 
Real-world response and survival rates were similar to clinical trials.
Real-world ORR, CR, PFS, and OS better with axi-cel compared to tisa-cel, although axi-cel with more toxicity. 
Chihara et al.31 
Retrospective cohort from Medicare claims data 
All CAR-T 551 31% of patients in this cohort were age ≥75 Not assessed Age ≥75:
mPFS 160 d
mOS 403 d
Age 70-74:
mPFS 379 d
mOS 603 d
Age 65-69:
mPFS 194
mOS 518
1 y PFS:
Age ≥75 34%
Age 70-74 52%
Age 65-69 43% 
Not assessed Not assessed Not assessed Not assessed Older patients, particularly those age ≥75, had significantly worse PFS and OS compared to younger patients as well as compared to clinical trials. 
Nastoupil et al.32 
Retrospective cohort from the US Lymphoma CAR-T Consortium 
Axi-cel 298 43% were ineligible for ZUMA-1 due to comorbidities ORR 82% (CR 64%) 1 y:
PFS 47%
OS 68%
PFS 34% for ZUMA-1 ineligible patients 
mDOR NR (median follow-up of 12.9 mo) 91% (7% grade 3+) 69% (31% grade 3+ 4.4%, 1 death due to HLH, 1 death due to ICANS Real-world outcomes and safety were comparable to ZUMA-1, although ECOG PS 2-4 had worse PFS, OS, and toxicities.
Patients ineligible for ZUMA-1 had inferior PFS and OS. 
Sano et al.33 
Retrospective cohort from the US Lymphoma CAR-T Consortium 
Axi-cel 272
30% were aged ≥65 
Not assessed Age ≥65
ORR 84% (CR 71%)
Age ≤65
ORR 82% (CR 51%) 
Age ≥65
mPFS 9.2 mo, mOS not assessable
Age ≤65
mPFS 7.4 mo, mOS 18.7 mo 
Not assessed Age ≥65
92% (7% grade 3+)
Age ≤65
91% (7% grade 3+) 
Age ≥65
78% (35% grade 3+)
Age ≤65
65% (31% grade 3+) 
2 deaths (1 in age ≥65 and 1 age <65) Rate of complete response was higher in patients aged ≥65 compared to age <65.
There was no difference in PFS, OS, and toxicities between patients aged <65 and ≥65. 
Bethge et al.34 
Retrospective cohort from the German Registry for Stem Cell Transplantation 
Axi-cel
Tisa-cel 
173 axi-cel
183 tisa-cel 
13% eligible for ZUMA-1
89% eligible for JULIET 
Axi-cel:
ORR 74% (CR 42%)
Tisa-cel:
ORR 53% (CR 32%) 
Axi-cel:
1 y PFS 35%, OS 55%
Tisa-cel:
1 y PFS 24%, OS 53% 
Not assessed Axi-cel:
81% (10% grade 3+)
Tisa-cel:
65% (13% grade 3+) 
Axi-cel:
44% (16% grade 3+)
Tisa-cel:
22% (7% grade 3+) 
Axi-cel:
2 y
10.4%
Tisa-cel:
2 y 3.5% 
Real-world ORR, CR, and OS were comparable to clinical trials, but real-world PFS was worse.
Higher rates of delayed infection-related NRM in real-world cohort. 
Riedell et al.35 
Retrospective cohort of 8 US centers
 
Axi-cel
Tisa-cel 
168 axi-cel
92 tisa-cel 
61% axi-cel ineligible for ZUMA-1
43% tisa-cel ineligible for JULIET 
Axi-cel:
ORR 52% (CR 44%)
Tisa-cel:
ORR 41% (CR 35%)
 
Axi-cel:
1 y PFS 42%, OS 62%
Tisa-cel:
1 y PFS 32%, OS 59% 
Axi-cel:
1 y DOR 70%
Tisa-cel:
1 y DOR 75% 
Axi-cel:
85% (9% grade 3+)
Tisa-cel:
39% (1% grade 3+)
mDOR NR for either cohort 
Axi-cel:
56% (38% grade 3+)
Tisa-cel:
11% (1% grade 3+) 
Axi-cel:
9%, 4 deaths from ICANS
Tisa-cel:
7%
 
Real-world outcomes were slightly lower compared to ZUMA-1 and JULIET, although safety outcomes were comparable.
Axi-cel and tisa-cel had comparable efficacy, although less CRS and ICANS with tisa-cel. 
Pasquini et al.36 
Postauthorization safety study through the CIBMTR registry 
Tisa-cel 155 Not assessed ORR 62% (CR 40%) 1 y:
EFS 52%
OS 77% 
1 y DOR 61% 45% (4.5% grade 3+) 18% (5.1% grade 3+) 1.2% Similar efficacy and improved safety compared to JULIET. 
Kittai et al.37 
Retrospective cohort from 4 centers 
Axi-cel
Tisa-cel 
94 axi-cel
36 tisa-cel 
57% of patients had high comorbidity based on CIRS score ORR 68% (CR 42%) mPFS 6.7 mo
mOS NR
1 y OS 60% 
Not assessed 79% 58% 12.3%, 2 deaths due to CRS, 1 death due to ICANS Worse ECOG PS and comorbidities by CIRS score were associated with worse survival.
Presence of more comorbidities was not associated with worse CRS or ICANS. 
Cook et al.38 
Meta-analysis of prospective and retrospective studies of CAR-T in primary and secondary CNS lymphoma 
Multiple CAR-T products 30 patients with primary CNS lymphoma (PCNSL)
98 patients with secondary CNS lymphoma (SCNSL) 
Not assessed PCNSL:
ORR 64% (CR 56%)
SCNSL:
ORR 57% (CR 47%) 
PCNSL:
6-mo PFS 37%
SCNSL:
6-mo PFS 37% 
PCNSL:
mDOR 9 mo
SCNSL:
mDOR 4.6 mo 
PCNSL:
70% (13% grade 3+)
SCNSL:
72% (11% grade 3+) 
PCNSL:
53% (18% grade 3+)
SCNSL:
48% (26% grade 3+) 
Not assessed Incidence of CRS and ICANS in real-world cohort of PCNSL and SCNSL is comparable to clinical trials.
Approximately one-third of patients with PCNSL and SCNSL had 6-mo complete responses, which could be durable with longer follow-up. 
Lin et al.50 
Single-center retrospective cohort of older adults who did and did not receive CAR-T 
Axi-cel and tisa-cel 24 older adults received CAR-T
18 older adults did not receive CAR-T
25 younger adults (age <65) received CAR-T 
Not assessed 100-d CR 51% (for 49 patients who received CAR-T) 6-mo PFS 48%, OS 71% (for 49 patients who received CAR-T)
Older adults who received CAR-T had a significantly lower risk of death (HR 0.31) compared to older adults who did not receive CAR-T
No difference in PFS or OS between older or younger adults who received CAR-T by chronological age, functional limitations, cognitive impairment, or comorbidity burden 
Not assessed 83% (8% grade 2±) 54% (25% grade 2±) 2 deaths in older adults after CAR-T Older adults who receive CAR-T have better postrelapse OS compared to patients who instead receive chemotherapy and/or supportive care.
PFS, OS, and rates of CRS and ICANS are not different between older and younger adults who receive CAR-T. 

AE, adverse event; CIBMTR, Center for International Blood and Marrow Transplant Research; ECOG PS, Eastern Cooperative Oncology Group Performance Status; NRM, nonrelapse mortality.

When considering BsAb vs CAR-T therapy in the third-line setting, cross-trial comparisons (Table 1) suggest lower response rates and PFS with BsAb therapies with shorter follow-up. In particular, patients with refractory disease have lower CR rates to BsAb therapies as compared to CAR-T: for glofitamab, CR rates are 34% with refractory as compared to 70% with nonrefractory disease,8  and for epcoritamab, CR rates are 30% with refractory vs 53% with nonrefractory disease.11  The lack of a costimulatory domain on current CD20xCD3 BsAb constructs may account for this observation, as cytotoxicity of BsAb therapy is dependent on innate immunity, whereas CAR-T constructs contain 4-1BB or CD28 costimulatory domains, which promote T-cell proliferation and persistence. From a logistic perspective, although CAR-T therapy requires significant upfront resources surrounding apheresis, CAR production, and intensive monitoring and toxicity management, current BsAb therapy strategies require ongoing dosing every several weeks for up to 12 cycles (glofitamab) or indefinitely until progression (epcoritamab), which may incur additional time and resource costs for patients as well as centers. Although CAR-T is considered cost-effective compared to other chemoimmunotherapy regimens and transplantation,39  cost-effectiveness studies between CAR-T and BsAb in the third-line setting are warranted. An additional advantage to sequencing CAR-T first is the demonstrated efficacy of BsAb therapies in patients with previous CAR-T exposure, whereas limited data have demonstrated efficacy of the converse strategy, with 1-year PFS and OS of 37% and 54%, respectively, in a registry cohort of 28 patients who received CAR-T after BsAb therapies.51  Further, there is theoretical concern about T-cell exhaustion post-BsAb therapy, which could limit the ability to apherese and manufacture an efficacious CAR T-cell product.40 

CAR-T eligibility

Given the curative potential of CAR-T among diverse patient cohorts, there is not a uniform definition for CAR-T eligibility, specifically for patients who may be considered too frail for CAR-T. Automatic exclusion of patients based on chronological age, presence of comorbidities, or cognitive and/or functional impairments is not advised as these characteristics do not consistently affect post–CAR-T outcomes.28,33,41,42  Further, transplant eligibility is not the same as CAR-T eligibility, as demonstrated in the PILOT trial of liso-cel in transplant-ineligible patients.20,43 

Based on patient cohorts with inferior outcomes after CAR-T (Table 2), we suggest that the following patients be referred to a multidisciplinary clinic with involvement by geriatrics, physical medicine and rehabilitation, physical therapy, occupational therapy, social work, and nutrition for comprehensive frailty assessment: age >70 to 75,31  Eastern Cooperative Oncology Group (ECOG) Performance Status of 2 or greater,25,28,29,32,37  multiple comorbidities per the Cumulative Illness Rating Scale (CIRS)- derived “Severe4” comorbidity index validated in CAR-T (CIRS score of 3 or higher in any of the respiratory, upper gastrointestinal, renal, or hepatic organ systems),37,44  significant weight loss and/or poor nutrition, and secondary CNS involvement and/or baseline cognitive impairment.41 

Although these characteristics have not yet been validated for determining CAR-T patient eligibility in routine clinical care, extrapolated evidence from the transplantation literature suggests that comprehensive frailty assessments in a dedicated multidisciplinary clinic may improve outcomes.45,46  Planning for functional optimization of at-risk patients before (“prehab”) and after (“rehab”) CAR-T, as well as discussion of bridging therapies and prophylactic steroids and/or anticytokine therapies prior to CAR-T infusion, should also occur.41 

After discussion with the patient, the decision was made to proceed with liso-cel CAR-T therapy, which was complicated by grade 2 CRS and grade 2 neurotoxicity, both of which completely resolved. One year after CAR-T therapy, he developed relapsed DLBCL in a single cervical lymph node, which was treated with radiation therapy resulting in a CR. Six months later, he developed progressive DLBCL.

Although CAR-T has the potential for cure in LBCL, two-thirds of patients will eventually relapse after CAR-T, and outcomes for these patients are dismal, with a median OS of only 5.2 months.47  There are not currently any data to inform which patients may preferentially benefit from BsAb first rather than CAR-T therapy to maximize survival. For the aforementioned patients who are known to have inferior survival after CAR-T, the potential benefits of BsAb therapies over CAR-T (Table 3) should be discussed, particularly the availability of off-the-shelf BsAb products and the generally lower rates of high-grade toxicities. These benefits should be placed in context of the unknown long-term curative potential of BsAb therapies, as well as the known activity of BsAb therapies in the post–CAR-T setting.

Table 3.

Pros and cons of CAR-T vs bispecific antibody therapies in third-line LBCL

CharacteristicCAR-T therapyBispecific antibody therapy
Curative potential Confirmed at 5 years of follow-up Data not yet mature; complete responses appear to be durable with short follow-up 
Administration Single infusion Repeated infusions required 
Off-the-shelf availability Not currently Yes 
Time to treatment initiation Several weeks required for product manufacturing Treatment can be started immediately 
Bridging therapy Optional, but may be needed in patients with aggressive disease Not needed 
Geographic availability Only at specialized, accredited centers Community settings 
Upfront commitment Substantial: hospitalization often required for administration Less than CAR-T: initial hospitalization required for dose escalation, although emerging data regarding safety of outpatient initiation 
Caregiver support required Substantial Less than CAR-T 
Time to best response ∼1 month ∼1.4 months 
T-cell requirements Dependent on T-cell health (manufacturing failures, bendamustine exposure) No costimulatory domain, so relies on innate immunity, which may be impaired by T-cell exhaustion 
Activity in CNS involvement CAR-T has demonstrated activity and comparable safety in primary and secondary CNS lymphoma Not yet studied 
Real-world data Comparable efficacy and safety in real-world cohorts as compared to clinical trial cohorts No real-world data regarding safety or efficacy 
Patient-reported outcomes Clinically meaningful longitudinal improvements in quality of life in the second- and third-line settings Data not yet published 
Cost-effectiveness Cost effective in the second and third lines Not yet studied 
Existing sequencing data No data for CAR-T after BsAb therapy Known efficacy and safety of BsAb therapy after CAR-T 
CharacteristicCAR-T therapyBispecific antibody therapy
Curative potential Confirmed at 5 years of follow-up Data not yet mature; complete responses appear to be durable with short follow-up 
Administration Single infusion Repeated infusions required 
Off-the-shelf availability Not currently Yes 
Time to treatment initiation Several weeks required for product manufacturing Treatment can be started immediately 
Bridging therapy Optional, but may be needed in patients with aggressive disease Not needed 
Geographic availability Only at specialized, accredited centers Community settings 
Upfront commitment Substantial: hospitalization often required for administration Less than CAR-T: initial hospitalization required for dose escalation, although emerging data regarding safety of outpatient initiation 
Caregiver support required Substantial Less than CAR-T 
Time to best response ∼1 month ∼1.4 months 
T-cell requirements Dependent on T-cell health (manufacturing failures, bendamustine exposure) No costimulatory domain, so relies on innate immunity, which may be impaired by T-cell exhaustion 
Activity in CNS involvement CAR-T has demonstrated activity and comparable safety in primary and secondary CNS lymphoma Not yet studied 
Real-world data Comparable efficacy and safety in real-world cohorts as compared to clinical trial cohorts No real-world data regarding safety or efficacy 
Patient-reported outcomes Clinically meaningful longitudinal improvements in quality of life in the second- and third-line settings Data not yet published 
Cost-effectiveness Cost effective in the second and third lines Not yet studied 
Existing sequencing data No data for CAR-T after BsAb therapy Known efficacy and safety of BsAb therapy after CAR-T 

It is critically important to assess patient preferences when deciding on sequencing of BsAb vs CAR-T therapies, as there are significant logistic disadvantages to CAR-T administration that may be burdensome to patients. CAR-T therapies are generally only available at large specialized cancer centers, which may require patients to travel great distances for consultation and apheresis, followed by prolonged stays away from home for CAR-T infusion and postinfusion monitoring.48  Further, although there are some centers that administer CAR-T therapies in the outpatient setting, most centers pursue inpatient administration for several weeks, which may be undesirable for patients and may lead to progressive frailty in older adults. BsAb therapies, on the other hand, will likely be available in the community setting and require much shorter inpatient stays for initial dose escalation, with formal studies ongoing to assess the safety of outpatient administration with premedication.8  A significant advantage of BsAb therapies is their off-the-shelf availability and capability for immediate treatment initiation, without requirement for apheresis and manufacturing, which may take several weeks for CAR-T therapies. This is particularly important for patients with florid and rapidly progressive LBCL, as interruptions in bridging therapies surrounding necessary washout periods before CAR-T apheresis and lymphodepleting chemotherapy may lead to significant disease progression during the manufacturing period and worse post–CAR-T outcomes and toxicities. In this vein, although CAR-T therapies have a time to best response of approximately 1 month, the time to best response of 1.4 months of glofitamab and epcoritamab may be preferable for patients with aggressive disease who may not be able to tolerate the additional several weeks of CAR-T manufacturing time. Finally, BsAb clinical trials in R/R LBCL included significant proportions of older patients (19% were age ≥75 years for epcoritamab and 54% were age ≥65 years for glofitamab),8,11  demonstrating efficacy and safety in this patient cohort. However, it is important to note that a subgroup analysis of patients aged ≥65 years in ZUMA-1 did not find any age-related differences in the efficacy or safety of axi-cel,49  which has been confirmed in real-world cohorts (Table 2). These data underscore the importance of a nuanced discussion with patients and their caregivers regarding the risks and benefits of CAR-T vs BsAb therapies.

The patient elected to enroll on a clinical trial of BsAb therapy mosunetuzumab in combination with polatuzumab vedotin. He received 8 cycles of combination therapy on trial and achieved a complete metabolic response, with his treatment course complicated by grade 1 CRS during the first 2 cycles and grade 3 neutropenia after cycle 5. His serial positron emission tomography scans at 12, 18, and 24 months after study enrollment continued to demonstrate ongoing remissions.

We propose a treatment algorithm for R/R LBCL that prioritizes CAR-T administration in all eligible patients in the second- and third-line settings given the curative potential and manageable toxicities of CAR-T in real-world cohorts (Figure 1).

Figure 1.

Our approach to sequencing CAR T-cell and bispecific antibody therapies in relapsed large B-cell lymphomas.

Figure 1.

Our approach to sequencing CAR T-cell and bispecific antibody therapies in relapsed large B-cell lymphomas.

Close modal

However, discussions with patients regarding their treatment preferences will be paramount as more long-term data on BsAb efficacy emerge in the literature. We recognize the limitations of cross-trial comparisons between CAR-T and BsAb therapies; prospective trials that compare the 2 strategies will be critical to treatment decision-making, particularly with risk stratification schema to study which patient cohorts may most benefit from CAR-T or BsAb therapies as the first in sequence. Ongoing trials studying specific CAR-T and BsAb sequencing strategies, such as SWOG 2114 and NCT04889716, which investigate various BsAb therapies as consolidation after commercial CAR-T, will also define optimal sequencing paradigms.52  Finally, efforts to increase the accessibility and safety of CAR-T delivery in community settings that are closer in proximity to patients and their caregivers will be vital in improving patient-centered outcomes.

Ajay Major: no competing financial interests to declare.

Manali Kamdar declares research support from Novartis; consultancy from AbbVie, AstraZeneca, Celgene/Bristol-Myers Squibb, Adaptive Biotechnologies, ADC Therapeutics, Beigene, Genentech, Impact Bio, Syncopation, and Caribou Biosciences; speaker's bureau from SeaGen; and data monitoring committee from Celgene and Genentech.

Ajay Major: Nothing to disclose.

Manali Kamdar: Nothing to disclose.

1.
Neelapu
SS
,
Jacobson
CA
,
Ghobadi
A
, et al.
5-Year follow-up supports curative potential of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1) [published online February 23, 2023]
.
Blood
.
doi:10.1182/blood.2022018893
.
2.
Chong
EA
,
Ruella
M
,
Schuster
SJ
;
Lymphoma Program Investigators at the University of Pennsylvania
.
Five-year outcomes for refractory B-cell lymphomas with CAR T-cell therapy
.
N Engl J Med
.
2021
;
384
(
7
):
673
-
674
.
doi:10.1056/NEJMc2030164
.
3.
Jain
T
,
Olson
TS
,
Locke
FL
.
How I treat cytopenias after CAR T-cell therapy
.
Blood
.
2023
;
141
(
20
):
2460
-
2469
.
doi:10.1182/blood.2022017415
.
4.
Maloney
DG
,
Kuruvilla
J
,
Liu
FF
, et al.
Matching-adjusted indirect treatment comparison of liso-cel versus axi-cel in relapsed or refractory large B cell lymphoma
.
J Hematol Oncol
.
2021
;
14
(
1
):
140
.
doi:10.1186/s13045-021-01144-9
.
5.
Neelapu
SS
,
Locke
FL
,
Bartlett
NL
, et al.
Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma
.
N Engl J Med
.
2017
;
377
(
26
):
2531
-
2544
.
doi:10.1056/NEJMoa1707447
.
6.
Schuster
SJ
,
Tam
CS
,
Borchmann
P
, et al.
Long-term clinical outcomes of tisagenlecleucel in patients with relapsed or refractory aggressive B-cell lymphomas (JULIET): a multicentre, open-label, single-arm, phase 2 study
.
Lancet Oncol
.
2021
;
22
(
10
):
1403
-
1415
.
doi:10.1016/S1470-2045(21)00375-2
.
7.
Abramson
JS
,
Palomba
ML
,
Gordon
LI
, et al.
Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study
.
Lancet
.
2020
;
396
(
10254
):
839
-
852
.
doi:10.1016/S0140-6736(20)31366-0
.
8.
Dickinson
MJ
,
Carlo-Stella
C
,
Morschhauser
F
, et al.
Glofitamab for relapsed or refractory diffuse large b-cell lymphoma
.
N Engl J Med
.
2022
;
387
(
24
):
2220
-
2231
.
doi:10.1056/NEJMoa2206913
.
9.
Hutchings
M
,
Carlo-Stella
C
,
Morschhauser
F
, et al.
Relapse is uncommon in patients with large B-cell lymphoma who are in complete remission at the end of fixed-course glofitamab treatment
.
Blood
.
2022
;
140
(
suppl 1
):
1062
-
1064
.
doi:10.1182/blood-2022-157554
.
10.
Hutchings
M
,
Mous
R
,
Clausen
MR
, et al.
Dose escalation of subcutaneous epcoritamab in patients with relapsed or refractory B-cell non-Hodgkin lymphoma: an open-label, phase ½ study
.
Lancet
.
2021
;
398
(
10306
):
1157
-
1169
.
doi:10.1016/S0140-6736(21)00889-8
.
11.
Thieblemont
C
,
Phillips
T
,
Ghesquieres
H
, et al.
Epcoritamab, a novel, subcutaneous CD3xCD20 bispecific T-cell-engaging antibody, in relapsed or refractory large B-cell lymphoma: dose expansion in a phase I/II trial
.
J Clin Oncol
.
2023
;
41
(
12
):
2238
-
2247
.
doi:10.1200/JCO.22.01725
.
12.
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
.
doi:10.1200/JCO.21.00931
.
13.
Bartlett
NL
,
Assouline
S
,
Giri
P
, et al.
Mosunetuzumab monotherapy is active and tolerable in patients with relapsed/refractory diffuse large B-cell lymphoma [published online April 17, 2023]
.
Blood Adv
.
doi:10.1182/bloodadvances.2022009260
.
14.
Bannerji
R
,
Arnason
JE
,
Advani
RH
, et al.
Odronextamab, a human CD20 × CD3 bispecific antibody in patients with CD20-positive B-cell malignancies (ELM-1): results from the relapsed or refractory non-Hodgkin lymphoma cohort in a single-arm, multicentre, phase 1 trial
.
Lancet Haematol
.
2022
;
9
(
5
):
e327
-
e339
.
doi:10.1016/S2352-3026(22)00072-2
.
15.
van Imhoff
GW
,
McMillan
A
,
Matasar
MJ
, et al.
Ofatumumab versus rituximab salvage chemoimmunotherapy in relapsed or refractory diffuse large B-cell lymphoma: the ORCHARRD Study
.
J Clin Oncol
.
2017
;
35
(
5
):
544
-
551
.
doi:10.1200/JCO.2016.69.0198
.
16.
Locke
FL
,
Miklos
DB
,
Jacobson
CA
, et al
;
All ZUMA-7 Investigators and Contributing Kite Members
.
Axicabtagene ciloleucel as second-line therapy for large B-cell lymphoma
.
N Engl J Med
.
2022
;
386
(
7
):
640
-
654
.
doi:10.1056/NEJMoa2116133
.
17.
Westin
JR
,
Locke
FL
,
Dickinson
M
, et al.
Safety and efficacy of axicabtagene ciloleucel versus standard of care in patients 65 years of age or older with relapsed/refractory large B-cell lymphoma
.
Clin Cancer Res
.
2023
;
29
(
10
):
1894
-
1905
.
doi:10.1158/1078-0432.CCR-22-3136
.
18.
Westin
JR
,
Oluwole
OO
,
Kersten
MJ
, et al
;
ZUMA-7 Investigators
;
Kite Members
.
Survival with axicabtagene ciloleucel in large B-cell lymphoma
.
N Engl J Med
.
2023
;
389
(
2
):
148
-
157
.
doi:10.1056/NEJMoa2301665
.
19.
Abramson
JS
,
Solomon
SR
,
Arnason
J
, et al.
Lisocabtagene maraleucel as second-line therapy for large B-cell lymphoma: primary analysis of the phase 3 TRANSFORM study
.
Blood
.
2023
;
141
(
14
):
1675
-
1684
.
doi:10.1182/blood.2022018730
.
20.
Sehgal
A
,
Hoda
D
,
Riedell
PA
, et al.
Lisocabtagene maraleucel as second-line therapy in adults with relapsed or refractory large B-cell lymphoma who were not intended for haematopoietic stem cell transplantation (PILOT): an open-label, phase 2 study
.
Lancet Oncol
.
2022
;
23
(
8
):
1066
-
1077
.
doi:10.1016/S1470-2045(22)00339-4
.
21.
Olszewski
AJ
,
Budde
LE
,
Chavez
J
, et al.
Mosunetuzumab with polatuzumab vedotin is effective and has a manageable safety profile in patients aged <65 and ≥65 years with relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL) and ≥1 prior therapy: subgroup analysis of a phase Ib/II study
.
Blood
.
2022
;
140
(
suppl 1
):
3757
-
3759
.
doi:10.1182/blood-2022-159594
22.
Patel
K
,
Riedell
PA
,
Tilly
H
, et al.
A phase 1 study of plamotamab, an anti-CD20 x anti-CD3 bispecific antibody, in patients with relapsed/refractory non-Hodgkin's lymphoma: recommended dose safety/efficacy update and escalation exposure-response analysis
.
Blood
.
2022
;
140
(
suppl 1
):
9470
-
9472
.
doi:10.1182/blood-2022-159586
.
23.
Thieblemont
C
,
Clausen
MR
,
Sureda Balari
A
, et al.
Phase 3 trial (GCT3013-05) of epcoritamab versus standard of care in patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL)
.
J Clin Oncol
.
2021
;
39
(
15, suppl
):
TPS7569
.
doi:10.1200/jco.2021.39.15_suppl.tps7569
.
24.
Jacobson
CA
,
Hunter
BD
,
Redd
R
, et al.
Axicabtagene ciloleucel in the non-trial setting: outcomes and correlates of response, resistance, and toxicity
.
J Clin Oncol
.
2020
;
38
(
27
):
3095
-
3106
.
doi:10.1200/JCO.19.02103
.
25.
Jacobson
CA
,
Locke
FL
,
Ma
L
, et al.
Real-world evidence of axicabtagene ciloleucel for the treatment of large B cell lymphoma in the United States
.
Transplant Cell Ther
.
2022
;
28
(
9
):
581.e1
-
581.e8
.
doi:10.1016/j.jtct.2022.05.026
.
26.
Patrick
DL
,
Powers
A
,
Jun
MP
, et al.
Effect of lisocabtagene maraleucel on HRQoL and symptom severity in relapsed/refractory large B-cell lymphoma
.
Blood Adv
.
2021
;
5
(
8
):
2245
-
2255
.
doi:10.1182/bloodadvances.2020003503
.
27.
Johnson
PC
,
Dhawale
T
,
Newcomb
RA
, et al.
Longitudinal patient-reported outcomes in patients receiving chimeric antigen receptor T-cell therapy
.
Blood Adv
.
2023
;
7
(
14
):
3541
-
3550
.
doi:10.1182/bloodadvances.2022009117
.
28.
Locke
FL
,
Jacobson
C
,
Ma
L
, et al.
Real-world outcomes of axicabtagene ciloleucel (axi-cel) for the treatment of large B-cell lymphoma (LBCL): impact of age and specific organ dysfunction
.
Blood
.
2021
;
138
(
suppl 1
):
530
.
doi:10.1182/blood-2021-149679
.
29.
Landsburg
DJ
,
Frigault
M
,
Heim
M
, et al.
Real-world outcomes for patients with relapsed or refractory (R/R) aggressive B-cell non-Hodgkin's lymphoma (aBNHL) treated with commercial tisagenlecleucel: subgroup analyses from the Center for International Blood and Marrow Transplant Research (CIBMTR) Registry
.
Blood
.
2022
;
140
(
suppl 1
):
1584
-
1587
.
doi:10.1182/blood-2022-158822
.
30.
Bachy
E
,
Le Gouill
S
,
Di Blasi
R
, et al.
A real-world comparison of tisagenlecleucel and axicabtagene ciloleucel CAR T cells in relapsed or refractory diffuse large B cell lymphoma
.
Nat Med
.
2022
;
28
(
10
):
2145
-
2154
.
doi:10.1038/s41591-022-01969-y
.
31.
Chihara
D
,
Liao
L
,
Tkacz
J
, et al.
Real-world effectiveness and economic impact associated with chimeric antigen receptor T-cell therapy among older patients with relapsed/refractory diffuse large B-cell lymphoma in US
.
Blood
.
2022
;
140
(
suppl 1
):
2421
-
2423
.
doi:10.1182/blood-2022-163678
.
32.
Nastoupil
LJ
,
Jain
MD
,
Feng
L
, et al.
Standard-of-care axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma: results from the US Lymphoma CAR T Consortium
.
J Clin Oncol
.
2020
;
38
(
27
):
3119
-
3128
.
doi:10.1200/JCO.19.02104
.
33.
Sano
D
,
Lekakis
L
,
Feng
L
, et al.
Safety and efficacy of axicabtagene ciloleucel (AXI-CEL) in older patients: results from the US lymphoma CAR-T consortium
.
Hematol Oncol
.
2019
;
37
:
304
-
305
.
doi:10.1002/hon.113_2630
.
34.
Bethge
WA
,
Martus
P
,
Schmitt
M
, et al.
GLA/DRST real-world outcome analysis of CAR T-cell therapies for large B-cell lymphoma in Germany
.
Blood
.
2022
;
140
(
4
):
349
-
358
.
doi:10.1182/blood.2021015209
.
35.
Riedell
PA
,
Hwang
W-T
,
Nastoupil
L-J
, et al.
Patterns of use, outcomes, and resource utilization among recipients of commercial axicabtagene ciloleucel and tisagenlecleucel for relapsed/refractory aggressive B cell lymphomas
.
Transplant Cell Ther
.
2022
;
28
(
10
):
669
-
676
.
doi:10.1016/j.jtct.2022.07.011
.
36.
Pasquini
MC
,
Hu
Z-H
,
Curran
K
, et al.
Real-world evidence of tisagenlecleucel for pediatric acute lymphoblastic leukemia and non-Hodgkin lymphoma
.
Blood Adv
.
2020
;
4
(
21
):
5414
-
5424
.
doi:10.1182/bloodadvances.2020003092
.
37.
Kittai
AS
,
Huang
Y
,
Gordon
M
, et al.
Comorbidities predict inferior survival in patients receiving chimeric antigen receptor T cell therapy for diffuse large B cell lymphoma: a multicenter analysis
.
Transplant Cell Ther
.
2021
;
27
(
1
):
46
-
52
.
doi:10.1016/j.bbmt.2020.09.028
.
38.
Cook
MR
,
Dorris
CS
,
Makambi
KH
, et al.
Toxicity and efficacy of CAR T-cell therapy in primary and secondary CNS lymphoma: a meta-analysis of 128 patients
.
Blood Adv
.
2023
;
7
(
1
):
32
-
39
.
doi:10.1182/bloodadvances.2022008525
.
39.
Lin
JK
,
Muffly
LS
,
Spinner
MA
,
Barnes
JI
,
Owens
DK
,
Goldhaber-Fiebert
JD
.
Cost effectiveness of chimeric antigen receptor T-cell therapy in multiply relapsed or refractory adult large B-cell lymphoma
.
J Clin Oncol
.
2019
;
37
(
24
):
2105
-
2119
.
doi:10.1200/JCO.18.02079
.
40.
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
.
doi:10.1182/blood.2022015956
.
41.
Shouse
G
,
Danilov
AV
,
Artz
A
.
CAR T-Cell Therapy in the older person: indications and risks
.
Curr Oncol Rep
.
2022
;
24
(
9
):
1189
-
1199
.
doi:10.1007/s11912-022-01272-6
.
42.
Greenwell
IB
,
Dahi
PB
.
Transplant and chimeric antigen receptor T-cell therapies in elderly patients with lymphoma, a narrative review
.
Ann Lymphoma
.
2022
;
6
:
8
-
8
.
doi:10.21037/aol-22-7
.
43.
Vic
S
,
Lemoine
J
,
Armand
P
,
Lemonnier
F
,
Houot
R
.
Transplant-ineligible but chimeric antigen receptor T-cells eligible: a real and relevant population
.
Eur J Cancer
.
2022
;
175
:
246
-
253
.
doi:10.1016/j.ejca.2022.08.019
.
44.
Shouse
G
,
Kaempf
A
,
Gordon
MJ
, et al.
A validated composite comorbidity index predicts outcomes of CAR T-cell therapy in patients with diffuse large B-cell lymphoma
.
Blood Adv
.
2023
;
7
(
14
):
3516
-
3529
.
doi:10.1182/bloodadvances.2022009309
.
45.
Derman
BA
,
Kordas
K
,
Ridgeway
J
, et al.
Results from a multidisciplinary clinic guided by geriatric assessment before stem cell transplantation in older adults
.
Blood Adv
.
2019
;
3
(
22
):
3488
-
3498
.
doi:10.1182/bloodadvances.2019000790
.
46.
Olin
RL
,
Fretham
C
,
Pasquini
MC
, et al.
Geriatric assessment in older alloHCT recipients: association of functional and cognitive impairment with outcomes
.
Blood Adv
.
2020
;
4
(
12
):
2810
-
2820
.
doi:10.1182/bloodadvances.2020001719
.
47.
Di Blasi
R
,
Le Gouill
S
,
Bachy
E
, et al.
Outcomes of patients with aggressive B-cell lymphoma after failure of anti-CD19 CAR T-cell therapy: a DESCAR-T analysis
.
Blood
.
2022
;
140
(
24
):
2584
-
2593
.
doi:10.1182/blood.2022016945
.
48.
Gajra
A
,
Zalenski
A
,
Sannareddy
A
,
Jeune-Smith
Y
,
Kapinos
K
,
Kansagra
A
.
Barriers to chimeric antigen receptor T-cell (CAR-T) therapies in clinical practice
.
Pharmaceut Med
.
2022
;
36
(
3
):
163
-
171
.
doi:10.1007/s40290-022-00428-w
.
49.
Neelapu
SS
,
Jacobson
CA
,
Oluwole
OO
, et al.
Outcomes of older patients in ZUMA-1, a pivotal study of axicabtagene ciloleucel in refractory large B-cell lymphoma
.
Blood
.
2020
;
135
(
23
):
2106
-
2109
.
doi:10.1182/blood.2019004162
.
50.
Lin
RJ
,
Lobaugh
SM
,
Pennisi
M
, et al.
Impact and safety of chimeric antigen receptor T-cell therapy in older, vulnerable patients with relapsed/refractory large B-cell lymphoma
.
Haematologica
.
2021
;
106
(
1
):
255
-
258
.
doi:10.3324/haematol.2019.243246
.
51.
Crochet
G
,
Audrey
C
,
Bachy
E
, et al.
CAR T-cell therapy remain effective in patients with relapse/refractory B-cell Non-Hodgkin lymphoma after bispecific antibodies exposure: results of a Lysa study based on the DESCAR-T registry
.
Blood
.
2022
;
140
(
suppl 1
):
4639
-
4641
.
doi:10.1182/blood-2022-162594
.
52.
Hess
B
,
Li
H
,
Hossain
N
, et al.
SWOG 2114: a randomized phase II trial of consolidation therapy following CD19 CAR T-cell treatment for relapsed/refractory large B-cell or grade IIIB follicular lymphoma
.
Hematol Oncol
.
2023
;
41
(
suppl 2
):
839
-
840
.
doi:10.1002/hon.3166_ot19
.