• Bridging intensity is associated with distinct post–CAR-T hematopoietic reconstitution patterns.

  • Intensive CTX bridging predisposes for severe late cytopenias, infection susceptibility and increased need for supportive measures.

Abstract

Chimeric antigen receptor (CAR) T-cell (CAR-T) therapy represents a major advance in the treatment of relapsed/refractory multiple myeloma (RRMM). However, the long time span from leukapheresis to actual CAR-T infusion often necessitates bridging therapies. Because of limited knowledge about the effects of bridging on post–CAR-T clinical course and outcomes, the selection of treatment options is challenging. In this multicenter international observational study, we explored the impact of bridging therapy on hematopoietic reconstitution in 158 patients with RRMM treated with B-cell maturation antigen (BCMA)–directed CAR-T therapy. Based on exposure to classical cytotoxic (CTX) chemotherapy, we classified bridging regimens as non-CTX, intermediate CTX (1-2 CTX agents), or intensive CTX (≥3 CTX agents or high-dose therapy with stem cell transplantation). We found associations between the number of CTX agents used and impaired post–CAR-T hematopoietic reconstitution, evident across hematopoietic cell lineages and particularly manifesting during the late post–CAR-T period. Intensive CTX bridging was associated with a prolonged time to neutrophil and platelet recovery, distinct patterns of hematopoietic recovery (eg, an intermittent phenotype characterized by a second drop), an increased susceptibility to severe infections and a significantly increased risk for severe late cytopenias in univariate and multivariate models. Taken together, these results highlight that bridging intensity distinctly shapes the trajectory of hematopoietic recovery after BCMA CAR-T therapy. Targeted and novel immunotherapies could provide alternatives for bridging, and high-risk patients may particularly benefit from enhanced monitoring, prophylaxis, and supportive care.

Chimeric antigen receptor (CAR) T-cell (CAR-T) therapy has led to a paradigm shift in the treatment of advanced B-cell malignancies.1,2 Safety and efficacy of B-cell maturation antigen (BCMA)–targeted idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel) in relapsed/refractory multiple myeloma (RRMM) have been demonstrated in clinical trials3,4 and real-world studies.5,6 Despite increasing experience and scientific advances, guiding patients through the pre– and post–CAR-T period remains challenging. The long time span from T-cell collection to actual CAR-T infusion often requires the application of bridging therapies. Considering the diverse therapeutic landscape in MM, ranging from conventional chemotherapy to targeted therapies and immunotherapies, there is still notable heterogeneity and limited evidence regarding the optimal selection of bridging agents.7 

Bridging therapies are critical for disease control or debulking. This is not only important to avoid disease-related complications and sequelae but also to mitigate the risk for post–CAR-T toxicities.7-9 Numerous studies have demonstrated associations between baseline tumor burden and neurological toxicities or cytokine release syndrome (CRS).10-12 At the same time, bridging therapies are considered a risk factor for post–CAR-T cytopenias.13-18 These were recently termed “immune effector cell–associated hematotoxicity” (ICAHT) and represent the most common high-grade adverse event of CAR-T therapy.18-21 Both, early and late cytopenias can substantially affect quality of life and morbidity.17,20,21 Moreover, profound and protracted neutropenia can predispose for infections, which are the main determinant of nonrelapse mortality.22-24 However, the negative impact of bridging on post–CAR-T hematopoietic reconstitution remains controversial, because there are also studies reporting no or contrary associations after CD19- and BCMA-directed CAR-T therapy.25,26 To date, studies on the clinical implications of different bridging approaches in RRMM are limited, and effects on the longitudinal recovery of different cell lineages have, to our knowledge, not been conclusively clarified.14,15 

Overall, it remains unclear to what extent bridging therapy influences cytopenias after CAR-T therapy in RRMM. To elucidate the impact of different intensities of cytotoxic (CTX) bridging on post–CAR-T hematopoietic recovery and cytopenia-associated complications, we performed a multicenter international retrospective study including 158 patients with RRMM who received an infusion of anti-BCMA CAR-Ts.

Patient selection and data collection

We analyzed 158 patients with RRMM who were consecutively treated with either cilta-cel (n = 22) or ide-cel (n = 136) at 3 university hospitals (Heidelberg University Hospital, University Hospital of Würzburg, and Dana-Farber Cancer Institute) until the end of May 2024 (Figure 1A). Data cutoff was 22 August 2024. Patients had received lymphodepletion with fludarabine and cyclophosphamide (n = 153) according to the manufacturer’s instructions,3,4 or bendamustine (n = 5) due to the fludarabine shortage27 (Table 1). Clinical data and institutional standard operating procedures have already been partially published.28 Data were extracted from the electronic patient management software and the original medical records whenever available. The total observation period for toxicities ranged from day 0 (day of CAR-T infusion) to day 90. The time period until day 30 was referred to as the early post–CAR-T period, and the period between day 31 and 90 was referred to as the late post–CAR-T period.28,29 Unless otherwise stated, laboratory values before lymphodepletion were collected with a leniency period of up to 5 days.28 Missing data on complications were mainly because of a limited follow-up time, external care with limited data access, or loss to follow-up.

Definition and characteristics of bridging therapy (BT) groups. (A) Flowchart providing information on cohorts, analysis workflow, BT concepts, and groups (non-CTX, intermediate CTX, and intensive CTX). (B) Distribution and allocation of patients to BT groups based on exposure to CTX agents during the bridging period. (C) Response before LD depending on the BT group. (D) Kaplan-Meier estimates of the probability of PFS in days since CAR-T infusion depending on the BT group. Median PFS in months and 95% CI are provided above the curves. HRs, 95% CI, and log-rank P value for group comparisons are provided below the curves. The number of patients at risk and the number of events per time point are listed in the table below. (E-G) Box plots showing the ANC (E), platelet count (F), and hemoglobin levels (G) before LD depending on the BT group. The number of measurements per group and the P value of the group comparisons are given above. The framed numbers are the median values. The dashed lines indicate the threshold values for CTC grade 3 and grade 4 events. CR, complete response; CTC, common terminology criteria (for adverse events); HD, high-dose therapy (with subsequent autologous stem cell transplantation); LD, lymphodepletion; MR, minimal response; NA, not available; NE, not evaluable (panel C); NE, not estimable (panel D); PD, progressive disease; PR, partial response; RTX, radiotherapy; SD, stable disease; VGPR, very good partial response.

Definition and characteristics of bridging therapy (BT) groups. (A) Flowchart providing information on cohorts, analysis workflow, BT concepts, and groups (non-CTX, intermediate CTX, and intensive CTX). (B) Distribution and allocation of patients to BT groups based on exposure to CTX agents during the bridging period. (C) Response before LD depending on the BT group. (D) Kaplan-Meier estimates of the probability of PFS in days since CAR-T infusion depending on the BT group. Median PFS in months and 95% CI are provided above the curves. HRs, 95% CI, and log-rank P value for group comparisons are provided below the curves. The number of patients at risk and the number of events per time point are listed in the table below. (E-G) Box plots showing the ANC (E), platelet count (F), and hemoglobin levels (G) before LD depending on the BT group. The number of measurements per group and the P value of the group comparisons are given above. The framed numbers are the median values. The dashed lines indicate the threshold values for CTC grade 3 and grade 4 events. CR, complete response; CTC, common terminology criteria (for adverse events); HD, high-dose therapy (with subsequent autologous stem cell transplantation); LD, lymphodepletion; MR, minimal response; NA, not available; NE, not evaluable (panel C); NE, not estimable (panel D); PD, progressive disease; PR, partial response; RTX, radiotherapy; SD, stable disease; VGPR, very good partial response.

Close modal
Table 1.

Patient and disease characteristics

Overall N = 158Non-CTX
n = 82
Intermediate CTX
n = 55
Intensive CTX
n = 21
P value
Age, y      
Median (Q1, Q364 (57, 70) 64 (59, 70) 65 (57, 71) 59 (50, 65) .043 
<70, n (%) 117 (74) 59 (72) 39 (71) 19 (90) .2 
≥70, n (%) 41 (26) 23 (28) 16 (29) 2 (9.5)  
Sex, n (%)     .7 
Male 103 (65) 54 (66) 37 (67) 12 (57)  
Female 55 (35) 28 (34) 18 (33) 9 (43)  
Race, n (%)     .7 
Asian 3 (1.9) 1 (1.2) 2 (3.6) 0 (0)  
Black or African American 6 (3.8) 4 (4.9) 1 (1.8) 1 (4.8)  
White 144 (91) 75 (91) 49 (89) 20 (95)  
Other 5 (3.2) 2 (2.4) 3 (5.5) 0 (0)  
Ethnicity, n (%)     .7 
Hispanic or Latino 3 (1.9) 1 (1.2) 2 (3.6) 0 (0)  
Not Hispanic or Latino 155 (98) 81 (99) 53 (96) 21 (100)  
ECOG PS score, n (%)     .6 
0-1 136 (95) 70 (93) 47 (96) 19 (100)  
2-3 7 (4.9) 5 (6.7) 2 (4.1) 0 (0)  
Unknown 15  
Time from initial diagnosis to CAR-T therapy, median (Q1, Q3), y 6.4 (4.1, 8.6) 6.3 (4.4, 8.7) 6.7 (4.1, 8.9) 6.3 (2.8, 8.3) .6 
ISS stage, n (%)     .4 
80 (59) 39 (55) 29 (63) 12 (63)  
II 45 (33) 28 (39) 11 (24) 6 (32)  
III 11 (8.1) 4 (5.6) 6 (13) 1 (5.3)  
Unknown 22 11  
R-ISS, n (%)     .014 
32 (24) 10 (14) 19 (41) 3 (15)  
II 97 (71) 56 (80) 25 (54) 16 (80)  
III 7 (5.1) 4 (5.7) 2 (4.3) 1 (5.0)  
Unknown 22 12  
Extramedullary disease, n (%)     .4 
No 90 (58) 50 (63) 28 (51) 12 (57)  
Yes 65 (42) 29 (37) 27 (49) 9 (43)  
Unknown  
Extraosseous disease, n (%)     .1 
No 117 (75) 63 (80) 42 (76) 12 (57)  
Yes 38 (25) 16 (20) 13 (24) 9 (43)  
Unknown  
Cytogenetics, n (%)     .2 
Standard risk 90 (60) 43 (57) 37 (70) 10 (50)  
High risk 59 (40) 33 (43) 16 (30) 10 (50)  
del(17p) 38 (26) 22 (29) 11 (21) 5 (25) .6 
t(4;14) 22 (15) 13 (17) 4 (7.5) 5 (25) .13 
t(14;16) 5 (3.4) 2 (2.6) 1 (1.9) 2 (10) .3 
+1q 68 (46) 32 (42) 28 (53) 8 (40) .4 
Unknown  
Bone marrow burden, n (%)     .6 
<50% 76 (79) 40 (77) 28 (85) 8 (73)  
≥50% 20 (21) 12 (23) 5 (15) 3 (27)  
Unknown 62 30 22 10  
Previous therapy lines, median (Q1, Q35 (5, 7) 5 (4, 7) 5 (5, 6) 6 (5, 7) .6 
Previous therapies, n (%)      
Autologous SCT 144 (91) 76 (93) 50 (91) 18 (86) .6 
Allogeneic SCT 12 (7.6) 4 (4.9) 4 (7.3) 4 (19) .12 
BCMA-targeted therapy 25 (16) 15 (18) 5 (9.1) 5 (24) .2 
Belantamab 24 (15) 15 (18) 4 (7) 5 (24) .1 
Bispecific T-cell engager 7 (4.4) 4 (4.9) 2 (3.6) 1 (4.8) > .9 
Talquetamab 5 (3.2) 4 (4.9) 1 (1.8) 0 (0) .14 
Teclistamab 2 (1.3) 0 (0) 1 (1.8) 1 (4.8)  
CAR-T therapy 1 (0.6) 0 (0) 1 (1.8)  0 (0) .5 
Triple-class refractory,§ n (%)     .2 
No 28 (18) 11 (14) 14 (25) 3 (14)  
Yes 129 (82) 70 (86) 41 (75) 18 (86)  
Unknown  
Penta-drug exposed,|| n (%)     .8 
No 31 (20) 16 (20) 12 (22) 3 (14)  
Yes 127 (80) 66 (80) 43 (78) 18 (86)  
Penta-drug refractory,|| n (%)     .5 
No 111 (72) 57 (71) 41 (76) 13 (62)  
Yes 44 (28) 23 (29) 13 (24) 8 (38)  
Unknown  
Center, n (%)     .004 
Germany 92 (58) 38 (46) 41 (75) 13 (62)  
United States 66 (42) 44 (54) 14 (25) 8 (38)  
CAR-T product, n (%)     .066 
Cilta-cel 22 (14) 11 (13) 11 (20) 0 (0)  
Ide-cel 136 (86) 71 (87) 44 (80) 21 (100)  
Out-of-specification CAR-T product, n (%)     .7 
No 151 (96) 78 (96) 52 (95) 21 (100)  
Yes 6 (3.8) 3 (3.7) 3 (5.5) 0 (0)  
Unknown  
Lymphodepletion, n (%)     .3 
Bendamustine 5 (3.2) 2 (2.4) 1 (1.8) 2 (9.5)  
Fludarabine/cyclophosphamide 153 (97) 80 (98) 54 (98) 19 (90)  
Vein-to-vein time,# median (Q1, Q3), d 53 (46, 63)∗∗  49 (43, 60) 58 (48, 69) 51 (48, 56) .022 
Overall N = 158Non-CTX
n = 82
Intermediate CTX
n = 55
Intensive CTX
n = 21
P value
Age, y      
Median (Q1, Q364 (57, 70) 64 (59, 70) 65 (57, 71) 59 (50, 65) .043 
<70, n (%) 117 (74) 59 (72) 39 (71) 19 (90) .2 
≥70, n (%) 41 (26) 23 (28) 16 (29) 2 (9.5)  
Sex, n (%)     .7 
Male 103 (65) 54 (66) 37 (67) 12 (57)  
Female 55 (35) 28 (34) 18 (33) 9 (43)  
Race, n (%)     .7 
Asian 3 (1.9) 1 (1.2) 2 (3.6) 0 (0)  
Black or African American 6 (3.8) 4 (4.9) 1 (1.8) 1 (4.8)  
White 144 (91) 75 (91) 49 (89) 20 (95)  
Other 5 (3.2) 2 (2.4) 3 (5.5) 0 (0)  
Ethnicity, n (%)     .7 
Hispanic or Latino 3 (1.9) 1 (1.2) 2 (3.6) 0 (0)  
Not Hispanic or Latino 155 (98) 81 (99) 53 (96) 21 (100)  
ECOG PS score, n (%)     .6 
0-1 136 (95) 70 (93) 47 (96) 19 (100)  
2-3 7 (4.9) 5 (6.7) 2 (4.1) 0 (0)  
Unknown 15  
Time from initial diagnosis to CAR-T therapy, median (Q1, Q3), y 6.4 (4.1, 8.6) 6.3 (4.4, 8.7) 6.7 (4.1, 8.9) 6.3 (2.8, 8.3) .6 
ISS stage, n (%)     .4 
80 (59) 39 (55) 29 (63) 12 (63)  
II 45 (33) 28 (39) 11 (24) 6 (32)  
III 11 (8.1) 4 (5.6) 6 (13) 1 (5.3)  
Unknown 22 11  
R-ISS, n (%)     .014 
32 (24) 10 (14) 19 (41) 3 (15)  
II 97 (71) 56 (80) 25 (54) 16 (80)  
III 7 (5.1) 4 (5.7) 2 (4.3) 1 (5.0)  
Unknown 22 12  
Extramedullary disease, n (%)     .4 
No 90 (58) 50 (63) 28 (51) 12 (57)  
Yes 65 (42) 29 (37) 27 (49) 9 (43)  
Unknown  
Extraosseous disease, n (%)     .1 
No 117 (75) 63 (80) 42 (76) 12 (57)  
Yes 38 (25) 16 (20) 13 (24) 9 (43)  
Unknown  
Cytogenetics, n (%)     .2 
Standard risk 90 (60) 43 (57) 37 (70) 10 (50)  
High risk 59 (40) 33 (43) 16 (30) 10 (50)  
del(17p) 38 (26) 22 (29) 11 (21) 5 (25) .6 
t(4;14) 22 (15) 13 (17) 4 (7.5) 5 (25) .13 
t(14;16) 5 (3.4) 2 (2.6) 1 (1.9) 2 (10) .3 
+1q 68 (46) 32 (42) 28 (53) 8 (40) .4 
Unknown  
Bone marrow burden, n (%)     .6 
<50% 76 (79) 40 (77) 28 (85) 8 (73)  
≥50% 20 (21) 12 (23) 5 (15) 3 (27)  
Unknown 62 30 22 10  
Previous therapy lines, median (Q1, Q35 (5, 7) 5 (4, 7) 5 (5, 6) 6 (5, 7) .6 
Previous therapies, n (%)      
Autologous SCT 144 (91) 76 (93) 50 (91) 18 (86) .6 
Allogeneic SCT 12 (7.6) 4 (4.9) 4 (7.3) 4 (19) .12 
BCMA-targeted therapy 25 (16) 15 (18) 5 (9.1) 5 (24) .2 
Belantamab 24 (15) 15 (18) 4 (7) 5 (24) .1 
Bispecific T-cell engager 7 (4.4) 4 (4.9) 2 (3.6) 1 (4.8) > .9 
Talquetamab 5 (3.2) 4 (4.9) 1 (1.8) 0 (0) .14 
Teclistamab 2 (1.3) 0 (0) 1 (1.8) 1 (4.8)  
CAR-T therapy 1 (0.6) 0 (0) 1 (1.8)  0 (0) .5 
Triple-class refractory,§ n (%)     .2 
No 28 (18) 11 (14) 14 (25) 3 (14)  
Yes 129 (82) 70 (86) 41 (75) 18 (86)  
Unknown  
Penta-drug exposed,|| n (%)     .8 
No 31 (20) 16 (20) 12 (22) 3 (14)  
Yes 127 (80) 66 (80) 43 (78) 18 (86)  
Penta-drug refractory,|| n (%)     .5 
No 111 (72) 57 (71) 41 (76) 13 (62)  
Yes 44 (28) 23 (29) 13 (24) 8 (38)  
Unknown  
Center, n (%)     .004 
Germany 92 (58) 38 (46) 41 (75) 13 (62)  
United States 66 (42) 44 (54) 14 (25) 8 (38)  
CAR-T product, n (%)     .066 
Cilta-cel 22 (14) 11 (13) 11 (20) 0 (0)  
Ide-cel 136 (86) 71 (87) 44 (80) 21 (100)  
Out-of-specification CAR-T product, n (%)     .7 
No 151 (96) 78 (96) 52 (95) 21 (100)  
Yes 6 (3.8) 3 (3.7) 3 (5.5) 0 (0)  
Unknown  
Lymphodepletion, n (%)     .3 
Bendamustine 5 (3.2) 2 (2.4) 1 (1.8) 2 (9.5)  
Fludarabine/cyclophosphamide 153 (97) 80 (98) 54 (98) 19 (90)  
Vein-to-vein time,# median (Q1, Q3), d 53 (46, 63)∗∗  49 (43, 60) 58 (48, 69) 51 (48, 56) .022 

Significant P values are set in boldface.

ECOG PS, Eastern Cooperative Oncology Group performance status; ISS, International Staging System; Q1, lower quartile; Q3, upper quartile; R-ISS, Revised International Staging System; SCT, stem cell transplant.

Determined before lymphodepletion.

Last bone marrow status determined within 90 days before CAR-T therapy.

Allogeneic anti-BCMA CAR-T therapy.

§

Refractory to an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 monoclonal antibody.

||

Exposed/refractory to lenalidomide, pomalidomide, bortezomib, carfilzomib, and daratumumab.

Refers to the final quality of the CAR-T product administered to the patient.

#

Refers to the time between (final) leukapheresis and CAR-T infusion.

∗∗

Vein-to-vein time (median [Q1, Q3]) depending on CAR-T product: 49 days (45, 58) for ide-cel, and 69 days (63, 84) for cilta-cel (P < .001).

Definition and classification of holding and bridging therapies

The term holding therapy refers to any antimyeloma treatment applied in the period before leukapheresis,30,31 provided that the time period between last exposure and final leukapheresis was <60 days. Bridging therapy was defined as any treatment applied between leukapheresis and lymphodepletion.7,13,30,31 Based on exposure to classical CTX chemotherapy, we differentiated between non-CTX (no CTX agents; eg, daratumumab/carfilzomib/dexamethasone), intermediate CTX (1-2 CTX agents; eg, carfilzomib/cyclophosphamide/dexamethasone, cyclophosphamide/etoposide/dexamethasone), and intensive CTX (≥3 CTX agents or high-dose therapy with autologous stem cell transplantation; eg, cisplatin, doxorubicin, cyclophosphamide, and etoposide) approaches (Figure 1A-B; supplemental Tables 1 and 2). Patients without systemic treatment were classified as non-CTX. If 2 different regimens were applied, patients were assigned according to the most CTX treatment. An independent radiotherapy expert ruled out a relevant effect on bone marrow function when patients received radiotherapy alone or concurrently, which was therefore not an exclusion criterion for the further analysis.

Analysis of cytopenias and hematopoietic recovery

Cytopenias were graded according to Common Terminology Criteria for Adverse Events, version 5.0. Grade ≥3 cytopenias were defined as severe. Nadir values were based on the lowest documented cell count, regardless of the underlying cause.28 Only patients with repetitive measurements (≥2 time points) during the respective observation period were included,28 unless the criteria for severe cytopenia were met. For analysis of hematopoietic reconstitution over time, blood cell counts available closest in time to the following time points were recorded: day 0 (−2 days), day 7 (±3 days), day 14 (±3 days), day 21 (±3 days), day 28 (±3 days), day 35 (±3 days), day 42 (±3 days), day 50 (±5 days), day 63 (±7 days), and day 90 (±7 days). Following the approach described by Juluri et al,32 time to neutrophil recovery was defined as the time from the first absolute neutrophil count (ANC) drop of <1000/μL to 3 consecutive measurements obtained on different days with an ANC of ≥1000/μL. Time to platelet recovery was defined as the time from the first platelet drop of <50 × 109/L to 3 consecutive measurements with a platelet count of ≥50 × 109/L. The first measurement was counted as the day of recovery. In the few cases in which recovery occurred without available confirmatory measurements, these were counted as recovery events. The thresholds were derived from CTC grade ≥3 and selected based on case numbers and clinical relevance. The phenotypes of neutrophil recovery (quick, intermittent, and aplastic) were defined according to Rejeski et al.19 

Other classifications and grading systems

Treatment response was assessed by the investigators on the basis of the International Myeloma Working Group criteria,33 with the usual adjustments and limitations due to the retrospective design.5 An absence of response data was classified as “not available.” Patients with no measurable disease and no evidence of disease progression were classified as “not evaluable.” Disease stage according to the Revised International Staging System,34,35 drug refractoriness, and previous lines of therapy36,37 were determined according to international consensus. Extramedullary disease included bone-associated (paramedullary) and bone-independent soft tissue masses38; extraosseous disease referred exclusively to the latter form.38 High-risk cytogenetics was defined as presence of del(17p), t(4;14), and/or t(14;16).3,4 Chromosome 1q21 abnormalities (+1q) included 1q gain or amplification.39,40 CRS and immune effector cell–associated neurotoxicity syndrome (ICANS) were graded according to the American Society for Transplantation and Cellular Therapy.41 Infections were defined and graded as previously described.42,43 For the analysis of severe infections, only grade ≥3 events with an onset between CAR-T infusion and day 90 were included. An infection event required clinical, microbiological, or radiological evidence. Fever alone was not counted as an infection event.42 Nonrelapse mortality and causes of death were defined according to the literature.42,44 

Flow cytometric analysis of immune cell subsets

Quantification of immune cell subsets in the peripheral blood was performed by flow cytometry as part of routine diagnostics at each center. The immune status was determined ≤14 days before lymphodepletion.

Statistical analysis

R (version 4.4.1), GraphPad Prism (version 10.4.0), and BioRender were used for data analysis and visualization. Continuous variables were compared by nonparametric Mann-Whitney U or Kruskal-Wallis tests, and percentages by the Fisher exact or χ2 tests. Longitudinal blood cell counts were analyzed using a linear mixed model with random patient effect and group, time, and group-time interaction as fixed effects.28 Univariate and multivariate logistic regression analyses were performed to examine associations between severe cytopenias and selected binary or continuous variables. Associations with time to neutrophil and platelet recovery, time to severe infection, progression-free survival (PFS), and overall survival (OS) were analyzed by univariate or multivariate Cox proportional hazards regression. Multivariate models were based on a priori–selected variables reflecting differences in baseline characteristics and treatment center28,30 and established risk factors for post–CAR-T cytopenias.20,21,25 Kaplan-Meier curves were used to estimate PFS and OS and compared by log-rank test. Kaplan-Meier estimates were further used to obtain cumulative risk curves for time to neutrophil and platelet recovery. Competing risk analysis of recovery times was not performed because of the low incidence of concurrent events. In case of loss to follow-up, disease progression, start of a new systemic treatment, or death, patients were censored. For time to severe infection, we performed an additional competing risk analysis treating the latter 3 event types as competing risks and using the Aalen-Johansen estimator to calculate cumulative incidence functions. Groups were compared by the Gray test. Severe infection density until day 90 was defined as the mean number of events per 100 patient days considering the total days at risk.42 All P values were 2-sided and considered as statistically significant at P <.05.

This study was approved by institutional committees and boards (ethics committee of the Medical Faculty of Heidelberg University [S-096/2017] and the Dana-Farber Institutional Review Board [18-340]). Written informed consent was obtained from all patients. All procedures were conducted in accordance with the Declaration of Helsinki.

Patient and disease characteristics

Baseline characteristics for the total cohort (n = 158) are provided in Table 1. The median age was 64 years (interquartile range [IQR], 57-70), and 55 patients (35%) were female. Of 143 patients with available data, 7 (5%) had an Eastern Cooperative Oncology Group performance status score of ≥2, 11 of 136 (8%) had International Staging System stage III, 65 of 155 (42%) had extramedullary disease, 59 of 149 (40%) had high-risk cytogenetics, and 20 of 96 (21%) had a bone marrow burden of ≥50%. Patients had received a median of 5 previous therapy lines (IQR, 5-7). Triple-class and penta-drug refractory disease were found in 129 of 157 (82%) and 44 of 155 (28%) evaluable patients, respectively. The median vein-to-vein time was 53 days (IQR, 46-63).

Bridging therapy groups and response status

Of the total cohort, 82 patients (52%) were assigned to the non-CTX, 55 patients (35%) to the intermediate CTX, and 21 patients (13%) to the intensive CTX bridging therapy group (Figure 1A-B). Details on the administered agents and regimens are shown in supplemental Tables 1 and 2. Five patients had received radiotherapy during the bridging period, whereby the effect on the bone marrow reserve was considered negligible after assessment by an independent radiotherapy expert. Differences regarding the distribution of patient and disease characteristics are outlined in Table 1. Comparing the allocation with a treatment group between the holding and bridging periods at the individual patient level (supplemental Figure 1), we observed continuity of the previous treatment group in 79% (n = 65), 62% (n = 34), and 43% (n = 9) of patients in the non-CTX, intermediate CTX, and intensive CTX bridging groups, respectively. We found no significant differences in the distribution of treatment responses before leukapheresis or lymphodepletion according to bridging intensity (Figure 1C; supplemental Table 3).

Laboratory parameters before lymphodepletion

A comparison of laboratory parameters before lymphodepletion is provided in supplemental Table 4. The intensive CTX bridging group showed a higher frequency of patients with increased LDH (57%) compared with the non-CTX (31%) and intermediate CTX (11%) groups (P < .001). Moreover, we observed higher median ferritin levels in the intensive CTX group (369 ng/mL) compared with the others (191 ng/mL and 141 ng/mL, respectively; P = .13). Significant differences were seen in CD3+ and CD4+ T-cell and CD19+ B-cell counts, with higher levels in the non-CTX group than in the intermediate CTX group.

ANC, platelet, and hemoglobin levels before lymphodepletion are illustrated in Figure 1E-G. CTC grades of cytopenias are given in supplemental Tables 5 and 6. Patients in the intermediate CTX and intensive CTX groups showed lower hemoglobin values (median: 10.3 g/dL and 10.2 g/dL, respectively) compared with the non-CTX group (median, 11.05 g/dL; P = .01 and P = .03, respectively), whereas no significant differences were found for neutrophils and platelets. However, the intensive CTX group exhibited a higher rate of severe cytopenia of any kind (33%) and of severe thrombocytopenia (19%) compared with the non-CTX (9.9% and 4.9%, respectively) and intermediate CTX groups (9.4% and 1.8%, respectively; P = .02 and P = .03, respectively).

CRS and ICANS

Most patients developed CRS (n = 134 [85%]), including 2 grade 3 events (1.3%). Patients in the intermediate CTX bridging group showed a significantly increased rate of CRS grade ≥2 events compared with all others (51% vs 26%; odds ratio [OR], 2.92; 95% confidence interval [CI], 1.50-5.85; P = .002). ICANS affected 16 patients (10%), with 4 grade ≥3 cases (2.5%) and an even distribution of ICANS cases across groups. Further information on safety is given in supplemental Table 7.

Efficacy of CAR-T therapy

Of the total cohort (n = 158), 124 patients (78%) achieved a partial response or better, and 55 patients (35%) had a complete response or better. No significant associations between best overall response and bridging groups were found (supplemental Table 3). With a median follow-up of 9.8 months (95% CI, 8.2-11.2), median PFS was 10.6 months (95% CI, 8.6-16.3), and median OS was not reached (supplemental Figure 2A-B). The intensive CTX group showed a significantly inferior PFS than the non-CTX group (hazard ratio [HR], 2.49; 95% CI, 1.27-4.87; P = .008; Figure 1D). Moreover, OS tended to be shorter among patients with intensive CTX bridging, albeit not significant (HR, 2.04; 95% CI, 0.77-5.40; P = .15; supplemental Figure 2C).

Intensive CTX bridging is associated with impaired post–CAR-T neutrophil recovery

The analysis of post–CAR-T ANC levels over time for the 3 bridging groups showed a lymphodepletion-associated drop, followed by a recovery (Figure 2A). Intensive CTX bridging was associated with a second drop, coinciding with the late post–CAR-T period. Average ANC levels across time points were significantly different between the non-CTX and intensive CTX groups (P = .01), and a global trend toward lower ANC levels was seen for the intermediate CTX group (P = .09). Moreover, there was a significant interaction between group and time points (P < .001), indicating different longitudinal recovery patterns (supplemental Table 8).

Figure 2.

Recovery of neutrophils after CAR-T therapy depending on the BT group. (A) Median ANC over time depending on the BT group (non-CTX, intermediate CTX, and intensive CTX). Measured events per group and time point are provided in the table below. P values are shown to the right of the table and refer to comparisons of groups across time points and interactions between group and time points. Further statistical analyses using a linear mixed model are summarized in supplemental Table 8. Baseline, before LD. day 0, day of CAR-T infusion. (B) Time to neutrophil recovery depending on the BT group. The dashed lines indicate the median values. HRs, 95% CIs, and P values for group comparisons are provided below the curves. The number of patients at risk per time point are listed in the table below. (C-D) Box plots showing the ANC nadir values during the early (C) and late (D) post–CAR-T period depending on the BT group. The number of measurements per group and the P value of the group comparisons are given above. The framed numbers are the median values. The dashed lines indicate the threshold values for CTC grade 3 and grade 4 events. (E) Distribution of neutrophil recovery phenotypes (quick [light green], intermittent [yellow], and aplastic [dark red]) depending on the BT group. The P value of the group comparison is shown above. (F) Cumulative incidence curves (day 0-90) showing the time to (first) severe infection (grade ≥3) by BT group. The P value based on the Gray test and the number of patients at risk per group is provided below the curves. (G) Comparison of severe infection densities per 100 patient days for the first 90 days after CAR-T infusion between the BT groups.

Figure 2.

Recovery of neutrophils after CAR-T therapy depending on the BT group. (A) Median ANC over time depending on the BT group (non-CTX, intermediate CTX, and intensive CTX). Measured events per group and time point are provided in the table below. P values are shown to the right of the table and refer to comparisons of groups across time points and interactions between group and time points. Further statistical analyses using a linear mixed model are summarized in supplemental Table 8. Baseline, before LD. day 0, day of CAR-T infusion. (B) Time to neutrophil recovery depending on the BT group. The dashed lines indicate the median values. HRs, 95% CIs, and P values for group comparisons are provided below the curves. The number of patients at risk per time point are listed in the table below. (C-D) Box plots showing the ANC nadir values during the early (C) and late (D) post–CAR-T period depending on the BT group. The number of measurements per group and the P value of the group comparisons are given above. The framed numbers are the median values. The dashed lines indicate the threshold values for CTC grade 3 and grade 4 events. (E) Distribution of neutrophil recovery phenotypes (quick [light green], intermittent [yellow], and aplastic [dark red]) depending on the BT group. The P value of the group comparison is shown above. (F) Cumulative incidence curves (day 0-90) showing the time to (first) severe infection (grade ≥3) by BT group. The P value based on the Gray test and the number of patients at risk per group is provided below the curves. (G) Comparison of severe infection densities per 100 patient days for the first 90 days after CAR-T infusion between the BT groups.

Close modal

The median time to neutrophil recovery after the first ANC drop of <1000/μL was 21 days (95% CI, 15-42) in the non-CTX group, 21 days (95% CI, 16-38) in the intermediate CTX group, and 63 days (95% CI, 29 to not estimable) in the intensive CTX group (Figure 2B). Patients in the latter group showed a significantly prolonged neutrophil recovery compared with all others (HR, 0.49; P = .04; supplemental Table 9).

Moreover, we observed significant differences between the 3 groups when comparing early and late ANC nadir values (Figure 2C-D). The univariate analysis showed significant associations between CTX bridging and severe early neutropenia (OR, 3.52; P = .005), and intensive CTX bridging and severe late neutropenia (OR, 2.89; P = .05; supplemental Tables 10 and 11).

The 3 bridging groups showed further differences in the distribution of neutrophil recovery phenotypes (P = .02) as defined by Rejeski et al19 (Figure 2E; supplemental Table 7), with an increasing proportion of intermittent recovery in parallel with cytotoxicity, and the highest proportion of aplastic phenotypes in the intensive CTX group. We observed a trend toward increasing rates of granulocyte-colony stimulating factor administrations during the early (46% vs 55% vs 70%; P = .3) and late (23% vs 34% vs 40%; P = .3) post–CAR-T periods across the 3 groups (supplemental Table 7). The recovery of leukocytes and lymphocytes is illustrated in supplemental Figure 3.

Intensive CTX bridging is associated with an increased susceptibility to severe infections

Severe infections affected 16 patients (20%) in the non-CTX group, 11 patients (20%) in the intermediate CTX group, and 8 patients (38%) in the intensive CTX group. In a univariate cause-specific Cox regression analysis, with the non-CTX group as reference, intensive CTX bridging showed a nearly significant association with the time to severe infection (HR, 2.32; 95% CI, 0.99-5.43; P = .05), whereas no association was observed for the intermediate CTX group (HR, 0.98; 95% CI, 0.45-2.11; P = .95). Adjusting for competing risks, the Aalan-Johansen estimator of the corresponding cumulative incidence curves is shown in Figure 2F. Moreover, the intensive CTX group was characterized by a higher severe infection density until day 90 (0.55) than the non-CTX (0.37) and intermediate CTX (0.31) groups (Figure 2G). Further details on the total of 47 severe infection events are provided in supplemental Figure 4.

Intensive CTX bridging is associated with severe thrombocytopenia

Longitudinal platelet recovery depending on bridging intensity is shown in Figure 3A. We observed a second, delayed decline of the platelet count, which was most pronounced in the intensive CTX group, with significant differences between the non-CTX and intensive CTX groups across time points (P = .006). Moreover, there was a significant interaction between group and time points (P < .001; supplemental Table 8).

Figure 3.

Recovery of PLTs after CAR-T therapy depending on the BT group. (A) Median PLT count over time depending on the BT group (non-CTX, intermediate CTX, and intensive CTX). Measured events per group and time point are provided in the table below. P values are shown to the right of the table and refer to comparisons of groups across time points and interactions between group and time points. Further statistical analyses using a linear mixed model are summarized in supplemental Table 8. Baseline, before LD. Day 0, day of CAR-T infusion. (B) Time to PLT recovery depending on the BT group. The dashed lines indicate the median values. HR, 95% CI, and P values for group comparisons are provided below the curves. The number of patients at risk per time point are listed in the table below. (C-D) Box plots showing the PLT count nadir values during the early (C) and late (D) post–CAR-T period depending on the BT group. The number of measurements per group and the P value of the group comparisons are given above. The framed numbers are the median values. The dashed lines indicate the threshold values for CTC grade 3 and grade 4 events. (E-G) The frequency of PLT transfusions during the late post–CAR-T period (E), TPO agonist administration (F), and autologous CD34+ stem cell boosts (G), depending on the BT group. The P value of the group comparison and, if indicated, the results of a contingency analysis, including ORs, 95% CI, and P value, are shown above. PLT, platelet; TPO, thrombopoietin receptor.

Figure 3.

Recovery of PLTs after CAR-T therapy depending on the BT group. (A) Median PLT count over time depending on the BT group (non-CTX, intermediate CTX, and intensive CTX). Measured events per group and time point are provided in the table below. P values are shown to the right of the table and refer to comparisons of groups across time points and interactions between group and time points. Further statistical analyses using a linear mixed model are summarized in supplemental Table 8. Baseline, before LD. Day 0, day of CAR-T infusion. (B) Time to PLT recovery depending on the BT group. The dashed lines indicate the median values. HR, 95% CI, and P values for group comparisons are provided below the curves. The number of patients at risk per time point are listed in the table below. (C-D) Box plots showing the PLT count nadir values during the early (C) and late (D) post–CAR-T period depending on the BT group. The number of measurements per group and the P value of the group comparisons are given above. The framed numbers are the median values. The dashed lines indicate the threshold values for CTC grade 3 and grade 4 events. (E-G) The frequency of PLT transfusions during the late post–CAR-T period (E), TPO agonist administration (F), and autologous CD34+ stem cell boosts (G), depending on the BT group. The P value of the group comparison and, if indicated, the results of a contingency analysis, including ORs, 95% CI, and P value, are shown above. PLT, platelet; TPO, thrombopoietin receptor.

Close modal

CTX bridging was significantly associated with a longer time to platelet recovery in univariate analysis (HR, 0.57; P = .05), with a median of 26 days (95% CI, 18-35) in the non-CTX group, 42 days (95% CI, 35-65) in the intermediate CTX group, and 55 days (95% CI, 21 to not estimable) in the intensive CTX group (Figure 3B; supplemental Table 12). Moreover, intensive CTX bridging was associated with lower platelet nadir values (Figure 3C-D) and an increased risk for severe thrombocytopenia during the early (OR, 4.11; P = .009) and late (OR, 9.06; P < .001) post–CAR-T periods (supplemental Table 13 and 14).

CTX bridging resulted in a significantly higher rate of platelet transfusions during the late post–CAR-T period compared with the non-CTX group (OR, 5.63; P = .008), and numerically higher rates of patients receiving thrombopoietin receptor agonists and stem cell boosts (Figure 3E-G; supplemental Table 7).

Intensive CTX bridging is associated with severe anemia during the late post–CAR-T period

Although the 3 bridging groups showed a similar recovery pattern of hemoglobin values during the early post–CAR-T period, there was a second decline among patients with intensive CTX bridging (Figure 4A). We observed significant differences across time points between the non-CTX and intensive CTX groups (P = .02), and time-group interaction analysis confirmed distinct temporal dynamics of hemoglobin recovery, driven by the intensive CTX group (P < .001; supplemental Table 8).

Figure 4.

Recovery of hemoglobin levels after CAR-T therapy depending on the BT group. (A) Median hemoglobin levels over time depending on the BT group (non-CTX, intermediate CTX, and intensive CTX). Measured events per group and time point are provided in the table below. P values are shown to the right of the table and refer to comparisons of groups across time points and interactions between group and time points. Further statistical analyses using a linear mixed model are summarized in supplemental Table 8. Baseline, before LD. Day 0, day of CAR-T infusion. (B-C) Box plots showing the hemoglobin nadir values during the early (B) and late (C) post–CAR-T period depending on the BT group. The number of measurements per group and the P value of the group comparisons are given above. The framed numbers are the median values. The dashed lines indicate the threshold values for CTC grade 3 events. (D-E) The frequency of RBC transfusions during the early (D) and late (E) post–CAR-T period depending on the BT group. The P value of the group comparison and, if indicated, the results of a contingency analysis, including OR, 95% CI, and P value, are shown above. RBC, red blood cell.

Figure 4.

Recovery of hemoglobin levels after CAR-T therapy depending on the BT group. (A) Median hemoglobin levels over time depending on the BT group (non-CTX, intermediate CTX, and intensive CTX). Measured events per group and time point are provided in the table below. P values are shown to the right of the table and refer to comparisons of groups across time points and interactions between group and time points. Further statistical analyses using a linear mixed model are summarized in supplemental Table 8. Baseline, before LD. Day 0, day of CAR-T infusion. (B-C) Box plots showing the hemoglobin nadir values during the early (B) and late (C) post–CAR-T period depending on the BT group. The number of measurements per group and the P value of the group comparisons are given above. The framed numbers are the median values. The dashed lines indicate the threshold values for CTC grade 3 events. (D-E) The frequency of RBC transfusions during the early (D) and late (E) post–CAR-T period depending on the BT group. The P value of the group comparison and, if indicated, the results of a contingency analysis, including OR, 95% CI, and P value, are shown above. RBC, red blood cell.

Close modal

Intensive CTX bridging was significantly associated with lower hemoglobin nadirs and severe anemia during the late (OR, 5.4; P = .003), but not early, post–CAR-T period (Figure 4B-C; supplemental Tables 15 and 16). Moreover, CTX bridging led to a significantly higher need for late red blood cell transfusions (OR, 4.16; P = .02; Figure 4E; supplemental Table 7).

Multivariate analysis identifies intensive CTX bridging as an independent risk factor

To further understand the impact of bridging intensity on post–CAR-T cytopenias, we performed multivariate modeling accounting for potential confounders and including established risk factors (Tables 2-4). Intensive CTX bridging was confirmed as an independent risk factor for severe early and late thrombocytopenia, severe late anemia, and a prolonged time to neutrophil recovery. Nearly significant associations were observed for severe late neutropenia and time to platelet recovery. Apart from bridging intensity, we identified CRS grade ≥2, preexisting grade ≥3 cytopenias, and disease progression before lymphodepletion as relevant risk markers of post–CAR-T cytopenias.

Table 2.

Multivariate analysis of severe neutropenia and neutrophil recovery during the post–CAR-T period

CharacteristicSevere neutropenia, early post–CAR-T period,
n = 129 (106 events)
Severe neutropenia, late post–CAR-T period
n = 114 (50 events)
Time to neutrophil recovery, n = 108 (83 events)
OR95% CIP valueOR95% CIP valueHR95% CIP value
Age, y (cont.) 1.02 0.95-1.08 .63 0.99 0.94-1.03 .56 0.98-1.03 .91 
Sex, female 2.45 0.78-9.07 .15 1.53 0.66-3.60 .33 0.82 0.50-1.35 .44 
Center, United States 0.77 0.09-17.1 .83 1.11 0.20-5.85 .9 0.91 0.33-2.50 .86 
Previous therapy lines, greater than the median (>5): yes 0.69 0.24-1.96 .49 1.86 0.83-4.23 .13 0.73 0.45-1.16 .18 
G-CSF prophylaxis, yes 0.25 0.01-1.96 .24 0.81 0.15-4.63 .81 0.91 0.30-2.75 .86 
PD before LD, yes 1.93 0.68-5.81 .22 2.33 1.02-5.52 .048 0.42 0.24-0.71 .001 
Grade ≥3 neutropenia, thrombocytopenia, and/or anemia before LD: yes 4.72 0.66-98.5 .18 1.02 0.28-3.68 .98 1.21 0.62-2.39 .58 
CRS grade ≥2, yes 4.67 1.30-22.9 .031 2.49 1.03-6.20 .045 0.89 0.55-1.44 .65 
BT: intensive CTX, yes 2.17 0.44-16.4 .38 3.52 0.92-15.1 .073 0.36 0.17-0.78 .01 
CharacteristicSevere neutropenia, early post–CAR-T period,
n = 129 (106 events)
Severe neutropenia, late post–CAR-T period
n = 114 (50 events)
Time to neutrophil recovery, n = 108 (83 events)
OR95% CIP valueOR95% CIP valueHR95% CIP value
Age, y (cont.) 1.02 0.95-1.08 .63 0.99 0.94-1.03 .56 0.98-1.03 .91 
Sex, female 2.45 0.78-9.07 .15 1.53 0.66-3.60 .33 0.82 0.50-1.35 .44 
Center, United States 0.77 0.09-17.1 .83 1.11 0.20-5.85 .9 0.91 0.33-2.50 .86 
Previous therapy lines, greater than the median (>5): yes 0.69 0.24-1.96 .49 1.86 0.83-4.23 .13 0.73 0.45-1.16 .18 
G-CSF prophylaxis, yes 0.25 0.01-1.96 .24 0.81 0.15-4.63 .81 0.91 0.30-2.75 .86 
PD before LD, yes 1.93 0.68-5.81 .22 2.33 1.02-5.52 .048 0.42 0.24-0.71 .001 
Grade ≥3 neutropenia, thrombocytopenia, and/or anemia before LD: yes 4.72 0.66-98.5 .18 1.02 0.28-3.68 .98 1.21 0.62-2.39 .58 
CRS grade ≥2, yes 4.67 1.30-22.9 .031 2.49 1.03-6.20 .045 0.89 0.55-1.44 .65 
BT: intensive CTX, yes 2.17 0.44-16.4 .38 3.52 0.92-15.1 .073 0.36 0.17-0.78 .01 

Significant P values are set in boldface.

BT, bridging therapy; cont., continuous; G-CSF, granulocyte-colony stimulating factor; LD, lymphodepletion; PD, progressive disease.

Table 3.

Multivariate analysis of severe thrombocytopenia and platelet recovery during the post–CAR-T period

CharacteristicSevere thrombocytopenia, early post–CAR-T period
n = 129 (62 events)
Severe thrombocytopenia, late post–CAR-T period
n = 116 (41 events)
Time to platelet recovery n = 64 (41 events)
OR95% CIP valueOR95% CIP valueHR95% CIP value
Age, y (cont.) 1.02 0.97-1.07 .45 1.04 0.99-1.10 .17 0.96 0.92-1.00 .068 
Sex, female 0.88 0.37-2.07 .77 1.14 0.44-2.93 .78 0.77 0.38-1.56 .47 
Center, United States 2.82 1.22-6.78 .017 1.07 0.40-2.75 .89 1.31 0.67-2.59 .43 
Previous therapy lines >5, yes 0.94 0.42-2.12 .88 1.8 0.73-4.54 .2 0.77 0.38-1.55 .47 
PD before LD, yes 2.87 1.29-6.63 .011 1.86 0.75-4.82 .19 0.61 0.29-1.29 .2 
Grade ≥3 neutropenia, thrombocytopenia, and/or anemia before LD: yes 3.3 0.99-13.1 .063 4.81 1.24-21.5 .027 0.38 0.16-0.92 .031 
CRS grade ≥2, yes 2.47 1.04-6.08 .043 1.23 0.46-3.20 .68 0.74 0.37-1.46 .38 
BT: intensive CTX, yes 6.32 1.81-26.6 .006 18.2 4.04-132 <.001 0.42 0.16-1.09 .073 
CharacteristicSevere thrombocytopenia, early post–CAR-T period
n = 129 (62 events)
Severe thrombocytopenia, late post–CAR-T period
n = 116 (41 events)
Time to platelet recovery n = 64 (41 events)
OR95% CIP valueOR95% CIP valueHR95% CIP value
Age, y (cont.) 1.02 0.97-1.07 .45 1.04 0.99-1.10 .17 0.96 0.92-1.00 .068 
Sex, female 0.88 0.37-2.07 .77 1.14 0.44-2.93 .78 0.77 0.38-1.56 .47 
Center, United States 2.82 1.22-6.78 .017 1.07 0.40-2.75 .89 1.31 0.67-2.59 .43 
Previous therapy lines >5, yes 0.94 0.42-2.12 .88 1.8 0.73-4.54 .2 0.77 0.38-1.55 .47 
PD before LD, yes 2.87 1.29-6.63 .011 1.86 0.75-4.82 .19 0.61 0.29-1.29 .2 
Grade ≥3 neutropenia, thrombocytopenia, and/or anemia before LD: yes 3.3 0.99-13.1 .063 4.81 1.24-21.5 .027 0.38 0.16-0.92 .031 
CRS grade ≥2, yes 2.47 1.04-6.08 .043 1.23 0.46-3.20 .68 0.74 0.37-1.46 .38 
BT: intensive CTX, yes 6.32 1.81-26.6 .006 18.2 4.04-132 <.001 0.42 0.16-1.09 .073 

Significant P values are set in boldface.

Table 4.

Multivariate analysis of severe anemia during the post–CAR-T period

CharacteristicSevere anemia, early post–CAR-T period
n = 129 (55 events)
Severe anemia, late post–CAR-T period
n = 115 (19 events)
OR95% CIP valueOR95% CIP value
Age, y (cont.) 1.01 0.96-1.06 .76    
Sex, female 2.84 1.25-6.70 .014    
Center, United States 1.81 0.80-4.15 .16 0.39 0.10-1.32 .15 
Previous therapy lines >5, yes 0.83 0.37-1.84 .65    
PD before LD, yes 2.12 0.96-4.83 .067 9.63 2.50-56.3 .003 
Grade ≥3 neutropenia, thrombocytopenia, and/or anemia before LD: yes 4.97 1.52-19.0 .011 4.54 1.02-21.3 .045 
CRS grade ≥2, yes 2.58 1.12-6.09 .027    
BT: intensive CTX, yes 0.86 0.24-2.89 .81 7.23 1.53-40.7 .015 
CharacteristicSevere anemia, early post–CAR-T period
n = 129 (55 events)
Severe anemia, late post–CAR-T period
n = 115 (19 events)
OR95% CIP valueOR95% CIP value
Age, y (cont.) 1.01 0.96-1.06 .76    
Sex, female 2.84 1.25-6.70 .014    
Center, United States 1.81 0.80-4.15 .16 0.39 0.10-1.32 .15 
Previous therapy lines >5, yes 0.83 0.37-1.84 .65    
PD before LD, yes 2.12 0.96-4.83 .067 9.63 2.50-56.3 .003 
Grade ≥3 neutropenia, thrombocytopenia, and/or anemia before LD: yes 4.97 1.52-19.0 .011 4.54 1.02-21.3 .045 
CRS grade ≥2, yes 2.58 1.12-6.09 .027    
BT: intensive CTX, yes 0.86 0.24-2.89 .81 7.23 1.53-40.7 .015 

Significant P values are set in boldface.

Here, we introduce a simple classification of bridging therapies, which can be easily transferred to everyday clinical practice and future studies. We found associations between CTX bridging intensity and temporal dynamics and phenotypes of post–CAR-T hematopoietic reconstitution. Intensive CTX bridging was associated with delayed recovery and a second drop in cell counts, leading to increased rates of severe cytopenias during the late post–CAR-T period.

Late cytopenias and the biphasic nature of hematopoietic recovery cannot be explained by lymphodepletion- or CRS-associated toxicity alone19,45 and seem to be driven by other mechanisms, such as redistribution and delayed immune effects of CAR-Ts,16,46 inhibition of hematopoietic stem cell maturation through paracrine effects47 or B-cell recovery related–perturbations of stromal derived factor 1 levels.45 Other explanations include cumulative bone marrow toxicity related to previous therapy lines and myeloma disease itself.18,21 Based on our findings, it can be hypothesized that intensive CTX bridging adds more damage to hematopoietic stem cells, inducing acute and latent long-term bone marrow injury.48 The reduced hematopoietic reserve associated with the latter form is masked under homeostatic conditions,48 but could undermine the later resistance to a “second hit” such as CAR-T–related inflammation and shifts in the bone marrow cytokine milieu. Of note, no differences between groups were observed for the time from initial diagnosis or the number of previous therapy lines, and a relevant proportion of patients in the intensive CTX group had received a less CTX holding therapy before. Although intensive CTX therapies were associated with a higher rate of severe cytopenias of any kind before lymphodepletion, differences regarding baseline cell counts were discrete. It is therefore noteworthy that differences regarding recovery patterns were most pronounced during the late post–CAR-T period, and that CTX bridging intensity was associated with increased levels of supportive care, including transfusions, growth factors, and stem cell boosts.

There is accumulating evidence for negative effects of bridging therapies on hematopoietic recovery after anti-BCMA-14,15 and anti-CD19 CAR-T therapy,13,16 and bridging therapies were taken up as a risk factor for ICAHT in the current consensus guidelines.18 Moreover, studies on bridging in RRMM have mainly demonstrated associations between alkylator-based regimens and inferior post–CAR-T outcomes,14,15 which is consistent with our findings. However, there are also studies that question the negative effects of bridging therapies on hematopoietic recovery. Logue et al observed an association between more time from last bridging and any grade ≥3 cytopenia on day 30 after ide-cel,25 and Xia et al reported an increased probability for cytopenias after CD19 CAR-T in subgroups with lower bridging rates.26 Moreover, debulking appears to be critical for protection from other immune-associated toxicities and favorable outcomes.10-13,49 Overall, there seems to be a fragile balance for the use of CTX bridging therapies between disease control, potentially leading to better outcomes and facilitating recovery of the hematopoietic niche,26 and impaired reconstitution by directly damaging the hematopoietic system, resulting in transfusion dependency and other complications.

The intensive CTX group was characterized by an increased tendency to severe infections. This could be explained by the impaired post–CAR-T recovery of immune cells but also by preceding negative effects of CTX chemotherapy on the humoral and cellular immune function and regulation,50,51 damage to mucosal barriers50,52 and endothelial dysfunction.53,54 

The major limitation of this study is the retrospective design. In addition, the cilta-cel cohort was characterized by limited patient numbers, a longer vein-to-vein time, and a shorter follow-up period. Another limitation arises from the fact that bridging groups showed differences regarding baseline characteristics and outcomes. Moreover, the results have to be interpreted under consideration of the multifactorial genesis of ICAHT.21 Aggressive disease manifestations are often countered by more intensive treatments, and differences in disease burden and dynamics before and after CAR-T infusion might be closely linked to increased toxicity. This makes it difficult to draw final conclusions regarding disease-, CAR-T–, and bridging-related effects. However, we aimed to address this point and adjust for potential confounders by including multivariate models and different end points.

In summary, our data underscore the relevance of intensive bridging as a risk factor for post–CAR-T cytopenias. In combination with clinical scores,19,28,55 bridging intensity should be considered for risk stratification before BCMA CAR-T therapy and risk-adapted planning and implementation of post–CAR-T clinical care under consideration of current guidelines.18 High-risk patients could benefit from alternative lymphodepletion regimens27,56,57; more frequent follow-ups; intensified prophylaxis; and supportive measures, including growth factors, transfusions, and immunoglobulin replacement. The availability of cryopreserved stem cells should be given special consideration when planning bridging and CAR-T therapy. If not available, alternatives to intensive bridging should be thoroughly evaluated, and a combined T-cell and stem cell collection approach may provide a solution for high-risk patients in the future.58 Although caution toward high-dose alkylators is also recommended in the current International Myeloma Working Group guidelines on treatment sequences,31 targeted therapies, and novel immunotherapies, including short-term use of bispecifics,59,60 appear to be promising in this context. The recent approval and increasing use of CAR-T therapies in earlier lines will open up new options for less hematotoxic bridging approaches and should be the preferred indication for cellular immunotherapies.

The authors thank all patients and the personnel at the participating centers who contributed to this work.

J.H.F. is funded through the International Myeloma Society and the Paula and Rodger Riney Foundation. A.S.S. has received funding from the National Cancer Institute (K08CA252174) and the Department of Defense (CA210827). M.S.R. has received research funding from the Dietmar Hopp Foundation.

Contribution: J.H.F., M.S.R., and S.S. conceptualized the study; J.H.F., T.H., and K.R. developed the methodology; J.H.F., X.Z., V.W., P.C., L.G., J.K., D.S., and K.H. performed the investigation; J.H.F. was responsible for analysis and visualization; M.S.R., S.S., L.R., A.S.S., O.N., E.K.M., C.S.M., M.J.F., N.W., H.G., A.S., M.H., M.S., C.M.-T., M.T., H.E., P.D., and N.C.M. were responsible for study supervision; J.H.F. wrote the original draft; M.S.R., K.R., and A.S.S. reviewed and edited the manuscript; M.S.R. was the study guarantor; and all authors read and approved the final manuscript.

Conflict-of-interest disclosure: J.H.F. reports an advisory role for Pfizer; honoraria from Bristol Myers Squibb (BMS) and Stemline Therapeutics; and travel and congress participation grants from Janssen-Cilag. K.R. reports research funding, honoraria, and travel support from Kite/Gilead; honoraria from Novartis and BMS/Celgene; travel support from Pierre-Fabre; and a consulting role for Kite/Gilead and BMS/Celgene. H.G. reports grants and/or provision of investigational medicinal products from Amgen, Array Biopharma/Pfizer, BMS/Celgene, Chugai, Dietmar Hopp Foundation, Janssen, Johns Hopkins University, Mundipharma GmbH, and Sanofi; research support from Amgen, BMS, Celgene, GlycoMimetics Inc, GlaxoSmithKline (GSK), Heidelberg Pharma, Hoffmann-La Roche, Karyopharm, Janssen, Incyte Corporation, Millenium Pharmaceuticals Inc, Molecular Partners, Merck Sharp & Dohme, MorphoSys AG, Pfizer, Sanofi, Takeda, and Novartis; has participated in advisory boards from Adaptive Biotechnology, Amgen, BMS, Janssen, and Sanofi; received honoraria from Amgen, BMS, Chugai, GSK, Janssen, Novartis, Sanofi, and Pfizer; and received support for attending meetings and/or travel support from Amgen, BMS, GSK, Janssen, Novartis, Sanofi, and Pfizer. A.S.S. reports consulting fees from Novartis. M.S.R. reports a consulting or advisory role for BMS, Amgen, GSK, Janssen, Sanofi, Pfizer, AbbVie, and Takeda; research funding from BMS, Janssen, Sanofi, and Heidelberg Pharma; travel support from BMS, Amgen, and Janssen; and honoraria from BMS, Janssen, AbbVie, and Sanofi. The remaining authors declare no competing financial interests.

Correspondence: Jan H. Frenking, Internal Medicine V, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; email: jan.frenking@med.uni-heidelberg.de; and Marc S. Raab, Internal Medicine V, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; email: marc.raab@med.uni-heidelberg.de.

1.
Gill
S
,
Brudno
JN
.
CAR T-cell therapy in hematologic malignancies: clinical role, toxicity, and unanswered questions
.
Am Soc Clin Oncol Educ Book
.
2021
;
41
:
e246
-
e265
.
2.
Zhang
X
,
Zhu
L
,
Zhang
H
,
Chen
S
,
Xiao
Y
.
CAR-T cell therapy in hematological malignancies: current opportunities and challenges
.
Front Immunol
.
2022
;
13
:
927153
.
3.
Munshi
NC
,
Anderson
LD
,
Shah
N
, et al
.
Idecabtagene vicleucel in relapsed and refractory multiple myeloma
.
N Engl J Med
.
2021
;
384
(
8
):
705
-
716
.
4.
Berdeja
JG
,
Madduri
D
,
Usmani
SZ
, et al
.
Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study
.
Lancet Lond Engl
.
2021
;
398
(
10297
):
314
-
324
.
5.
Hansen
DK
,
Sidana
S
,
Peres
LC
, et al
.
Idecabtagene vicleucel for relapsed/refractory multiple myeloma: real-world experience from the myeloma CAR T consortium
.
J Clin Oncol
.
2023
;
41
(
11
):
2087
-
2097
.
6.
Sidana
S
,
Patel
KK
,
Peres
LC
, et al
.
Safety and efficacy of standard of care ciltacabtagene autoleucel for relapsed/refractory multiple myeloma
.
Blood
.
2025
;
145
(
1
):
85
-
97
.
7.
Bal
S
,
Costa
LJ
.
Bridging treatment prior to chimeric antigen receptor T-cell therapy in multiple myeloma
.
Br J Haematol
.
2024
;
204
(
2
):
449
-
454
.
8.
Bhaskar
ST
,
Dholaria
BR
,
Sengsayadeth
SM
,
Savani
BN
,
Oluwole
OO
.
Role of bridging therapy during chimeric antigen receptor T cell therapy
.
eJHaem
.
2022
;
3
(
suppl 1
):
39
-
45
.
9.
Richter
J
.
Like a bridge over troubled water: keeping the myeloma down en route to CAR-T
.
Blood Cancer J
.
2024
;
14
(
1
):
64
.
10.
Cohen
AD
,
Parekh
S
,
Santomasso
BD
, et al
.
Incidence and management of CAR-T neurotoxicity in patients with multiple myeloma treated with ciltacabtagene autoleucel in CARTITUDE studies
.
Blood Cancer J
.
2022
;
12
(
2
):
32
-
39
.
11.
Villanueva
R
,
Hansen
DK
,
Tonseth
RP
, et al
.
High metabolic tumor volume is associated with higher toxicity and decreased efficacy of BCMA CAR-T cell therapy in multiple myeloma
.
Blood
.
2022
;
140
(
suppl 1
):
10402
-
10404
.
12.
Keijzer
K
,
de Boer
JW
,
van Doesum
JA
, et al
.
Reducing and controlling metabolic active tumor volume prior to CAR T-cell infusion can improve survival outcomes in patients with large B-cell lymphoma
.
Blood Cancer J
.
2024
;
14
(
1
):
41
-
49
.
13.
Roddie
C
,
Neill
L
,
Osborne
W
, et al
.
Effective bridging therapy can improve CD19 CAR-T outcomes while maintaining safety in patients with large B-cell lymphoma
.
Blood Adv
.
2023
;
7
(
12
):
2872
-
2883
.
14.
Afrough
A
,
Hashmi
H
,
Hansen
DK
, et al
.
Real-world impact of bridging therapy on outcomes of ide-cel for myeloma in the U.S. Myeloma Immunotherapy Consortium
.
Blood Cancer J
.
2024
;
14
(
1
):
63
-
65
.
15.
Zafar
A
,
Huang
CY
,
Lo
M
, et al
.
Intensity of cyclophosphamide-based bridging therapy before chimeric antigen receptor T cell therapy in myeloma
.
Transpl Cell Ther
.
2023
;
29
(
8
):
504.e1
-
504.e7
.
16.
Rejeski
K
,
Perez
A
,
Iacoboni
G
, et al
.
Severe hematotoxicity after CD19 CAR-T therapy is associated with suppressive immune dysregulation and limited CAR-T expansion
.
Sci Adv
.
2023
;
9
(
38
):
eadg3919
.
17.
Jain
T
,
Olson
TS
,
Locke
FL
.
How I treat cytopenias after CAR T-cell therapy
.
Blood
.
2023
;
141
(
20
):
2460
-
2469
.
18.
Rejeski
K
,
Subklewe
M
,
Aljurf
M
, et al
.
Immune effector cell-associated hematotoxicity (ICAHT): EHA/EBMT consensus grading and best practice recommendations
.
Blood
.
2023
;
142
(
10
):
865
-
877
.
19.
Rejeski
K
,
Perez
A
,
Sesques
P
, et al
.
CAR-HEMATOTOX: a model for CAR T-cell–related hematologic toxicity in relapsed/refractory large B-cell lymphoma
.
Blood
.
2021
;
138
(
24
):
2499
-
2513
.
20.
Rejeski
K
,
Wang
Y
,
Hansen
DK
, et al
.
Applying the EHA/EBMT grading for ICAHT after CAR-T: comparative incidence and association with infections and mortality
.
Blood Adv
.
2024
;
8
(
8
):
1857
-
1868
.
21.
Rejeski
K
,
Jain
MD
,
Shah
NN
,
Perales
MA
,
Subklewe
M
.
Immune effector cell-associated haematotoxicity after CAR T-cell therapy: from mechanism to management
.
Lancet Haematol
.
2024
;
11
(
6
):
e459
-
e470
.
22.
Cordas dos Santos
DM
,
Tix
T
,
Shouval
R
, et al
.
A systematic review and meta-analysis of nonrelapse mortality after CAR T cell therapy
.
Nat Med
.
2024
;
30
(
9
):
2667
-
2678
.
23.
Rejeski
K
,
Hansen
DK
,
Bansal
R
, et al
.
The CAR-HEMATOTOX score as a prognostic model of toxicity and response in patients receiving BCMA-directed CAR-T for relapsed/refractory multiple myeloma
.
J Hematol Oncol
.
2023
;
16
(
1
):
88
.
24.
Lemoine
J
,
Bachy
E
,
Cartron
G
, et al
.
Nonrelapse mortality after CAR T-cell therapy for large B-cell lymphoma: a LYSA study from the DESCAR-T registry
.
Blood Adv
.
2023
;
7
(
21
):
6589
-
6598
.
25.
Logue
JM
,
Peres
LC
,
Hashmi
H
, et al
.
Early cytopenias and infections after standard of care idecabtagene vicleucel in relapsed or refractory multiple myeloma
.
Blood Adv
.
2022
;
6
(
24
):
6109
-
6119
.
26.
Xia
Y
,
Zhang
J
,
Li
J
, et al
.
Cytopenias following anti-CD19 chimeric antigen receptor (CAR) T cell therapy: a systematic analysis for contributing factors
.
Ann Med
.
2022
;
54
(
1
):
2951
-
2965
.
27.
Sidana
S
,
Hosoya
H
,
Jensen
A
, et al
.
Bendamustine vs. fludarabine/cyclophosphamide lymphodepletion prior to BCMA CAR-T cell therapy in multiple myeloma
.
Blood Cancer J
.
2023
;
13
(
1
):
158
.
28.
Frenking
JH
,
Zhou
X
,
Wagner
V
, et al
.
EASIX-guided risk stratification for complications and outcome after CAR T-cell therapy with ide-cel in relapsed/refractory multiple myeloma
.
J Immunother Cancer
.
2024
;
12
(
10
):
e009220
.
29.
Wudhikarn
K
,
Perales
MA
.
Infectious complications, immune reconstitution, and infection prophylaxis after CD19 chimeric antigen receptor T-cell therapy
.
Bone Marrow Transpl
.
2022
;
57
(
10
):
1477
-
1488
.
30.
Bücklein
V
,
Perez
A
,
Rejeski
K
, et al
.
Inferior outcomes of EU versus US patients treated with CD19 CAR-T for relapsed/refractory large B-cell lymphoma: association with differences in tumor burden, systemic inflammation, bridging therapy utilization, and CAR-T product use [published correction appears in Hemasphere. 2023;7(11):e982]
.
HemaSphere
.
2023
;
7
(
8
):
e907
.
31.
Costa
LJ
,
Banerjee
R
,
Mian
H
, et al
.
International myeloma working group immunotherapy committee recommendation on sequencing immunotherapy for treatment of multiple myeloma
.
Leukemia
.
2025
;
39
(
3
):
543
-
554
.
32.
Juluri
KR
,
Wu
QV
,
Voutsinas
J
, et al
.
Severe cytokine release syndrome is associated with hematologic toxicity following CD19 CAR T-cell therapy
.
Blood Adv
.
2022
;
6
(
7
):
2055
-
2068
.
33.
Kumar
S
,
Paiva
B
,
Anderson
KC
, et al
.
International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma
.
Lancet Oncol
.
2016
;
17
(
8
):
e328
-
e346
.
34.
Greipp
PR
,
San Miguel
J
,
Durie
BGM
, et al
.
International Staging System for multiple myeloma [published correction appears in J Clin Oncol. 2005;23(25):6281]
.
J Clin Oncol
.
2005
;
23
(
15
):
3412
-
3420
.
35.
Palumbo
A
,
Avet-Loiseau
H
,
Oliva
S
, et al
.
Revised International Staging System for multiple myeloma: a report from International Myeloma Working Group
.
J Clin Oncol
.
2015
;
33
(
26
):
2863
-
2869
.
36.
Rajkumar
SV
,
Richardson
P
,
San Miguel
JF
.
Guidelines for determination of the number of prior lines of therapy in multiple myeloma
.
Blood
.
2015
;
126
(
7
):
921
-
922
.
37.
Rajkumar
SV
,
Harousseau
JL
,
Durie
B
, et al;
International Myeloma Workshop Consensus Panel 1
.
Consensus recommendations for the uniform reporting of clinical trials: report of the International Myeloma Workshop Consensus Panel 1
.
Blood
.
2011
;
117
(
18
):
4691
-
4695
.
38.
Bansal
R
,
Rakshit
S
,
Kumar
S
.
Extramedullary disease in multiple myeloma
.
Blood Cancer J
.
2021
;
11
(
9
):
161
.
39.
Schmidt
TM
,
Fonseca
R
,
Usmani
SZ
.
Chromosome 1q21 abnormalities in multiple myeloma
.
Blood Cancer J
.
2021
;
11
(
4
):
83
.
40.
D’Agostino
M
,
Cairns
DA
,
Lahuerta
JJ
, et al
.
Second revision of the International Staging System (R2-ISS) for overall survival in multiple myeloma: a European Myeloma Network (EMN) report within the HARMONY project
.
J Clin Oncol
.
2022
;
40
(
29
):
3406
-
3418
.
41.
Lee
DW
,
Santomasso
BD
,
Locke
FL
, et al
.
ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells
.
Biol Blood Marrow Transpl
.
2019
;
25
(
4
):
625
-
638
.
42.
Rejeski
K
,
Perez
A
,
Iacoboni
G
, et al
.
The CAR-HEMATOTOX risk-stratifies patients for severe infections and disease progression after CD19 CAR-T in R/R LBCL
.
J Immunother Cancer
.
2022
;
10
(
5
):
e004475
.
43.
Hill
JA
,
Li
D
,
Hay
KA
, et al
.
Infectious complications of CD19-targeted chimeric antigen receptor–modified T-cell immunotherapy
.
Blood
.
2018
;
131
(
1
):
121
-
130
.
44.
Mai
EK
,
Haas
EM
,
Lücke
S
, et al
.
A systematic classification of death causes in multiple myeloma
.
Blood Cancer J
.
2018
;
8
(
3
):
30
.
45.
Fried
S
,
Avigdor
A
,
Bielorai
B
, et al
.
Early and late hematologic toxicity following CD19 CAR-T cells
.
Bone Marrow Transpl
.
2019
;
54
(
10
):
1643
-
1650
.
46.
Wei
J
,
Liu
Y
,
Wang
C
, et al
.
The model of cytokine release syndrome in CAR T-cell treatment for B-cell non-Hodgkin lymphoma
.
Signal Transduct Target Ther
.
2020
;
5
(
1
):
134
.
47.
Palacios-Berraquero
ML
,
Rodriguez-Marquez
P
,
Calleja-Cervantes
ME
, et al
.
Molecular mechanisms promoting long-term cytopenia after BCMA CAR-T therapy in multiple myeloma
.
Blood Adv
.
2024
;
8
(
21
):
5479
-
5492
.
48.
Shao
L
,
Wang
Y
,
Chang
J
,
Luo
Y
,
Meng
A
,
Zhou
D
.
Hematopoietic stem cell senescence and cancer therapy-induced long-term bone marrow injury
.
Transl Cancer Res
.
2013
;
2
(
5
):
397
-
411
.
49.
Iacoboni
G
,
Sánchez-Salinas
MA
,
Rejeski
K
, et al
.
Efficacy and safety of bendamustine-containing bridging therapy in R/R LBCL patients receiving CD19 CAR T-cells
.
HemaSphere
.
2024
;
8
(
7
):
e86
.
50.
Vento
S
,
Cainelli
F
.
Infections in patients with cancer undergoing chemotherapy: aetiology, prevention, and treatment
.
Lancet Oncol
.
2003
;
4
(
10
):
595
-
604
.
51.
Sharma
A
,
Jasrotia
S
,
Kumar
A
.
Effects of chemotherapy on the immune system: implications for cancer treatment and patient outcomes
.
Naunyn Schmiedebergs Arch Pharmacol
.
2024
;
397
(
5
):
2551
-
2566
.
52.
Khan
SA
,
Wingard
JR
.
Infection and mucosal injury in cancer treatment
.
JNCI Monogr
.
2001
;
2001
(
29
):
31
-
36
.
53.
Luft
T
,
Dreger
P
,
Radujkovic
A
.
Endothelial cell dysfunction: a key determinant for the outcome of allogeneic stem cell transplantation
.
Bone Marrow Transpl
.
2021
;
56
(
10
):
2326
-
2335
.
54.
Terwoord
JD
,
Beyer
AM
,
Gutterman
DD
.
Endothelial dysfunction as a complication of anti-cancer therapy
.
Pharmacol Ther
.
2022
;
237
:
108116
.
55.
Rejeski
K
,
Hansen
DK
,
Cordas Dos Santos
DM
, et al
.
Pre-apheresis prediction of toxicity and response in patients receiving BCMA-directed CAR-T for relapsed/refractory multiple myeloma
.
Blood
.
2024
;
144
(
suppl 1
):
895
.
56.
Ghilardi
G
,
Chong
EA
,
Svoboda
J
, et al
.
Bendamustine is safe and effective for lymphodepletion before tisagenlecleucel in patients with refractory or relapsed large B-cell lymphomas
.
Ann Oncol
.
2022
;
33
(
9
):
916
-
928
.
57.
Ghilardi
G
,
Paruzzo
L
,
Svoboda
J
, et al
.
Bendamustine lymphodepletion before axicabtagene ciloleucel is safe and associates with reduced inflammatory cytokines
.
Blood Adv
.
2024
;
8
(
3
):
653
-
666
.
58.
Battram
AM
,
Oliver-Caldés
A
,
Suárez-Lledó
M
, et al
.
T cells isolated from G-CSF-treated multiple myeloma patients are suitable for the generation of BCMA-directed CAR-T cells
.
Mol Ther Methods Clin Dev
.
2022
;
26
:
207
-
223
.
59.
Fandrei
D
,
Seiffert
S
,
Rade
M
, et al
.
Bispecific antibodies as bridging to BCMA CAR-T cell therapy for relapsed/refractory multiple myeloma
.
Blood Cancer Discov
.
2025
;
6
(
1
):
38
-
54
.
60.
Dhakal
B
,
Akhtar
OS
,
Cowan
AJ
, et al
.
Talquetamab bridging: paving the way to B-cell maturation antigen (BCMA) CAR-T cell therapy in relapsed/refractory multiple myeloma (RRMM)
.
Blood
.
2024
;
144
(
suppl 1
):
931
.

Author notes

S.S. and M.S.R. contributed equally to this study.

The data sets generated and analyzed during this study are available upon reasonable request from the corresponding authors, Jan H. Frenking (jan.frenking@med.uni-heidelberg.de) and Marc S. Raab (marc.raab@med.uni-heidelberg.de).

The full-text version of this article contains a data supplement.

Supplemental data