Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 May 1.
Published in final edited form as: Leukemia. 2022 Sep 20;36(11):2669–2677. doi: 10.1038/s41375-022-01704-z

Impact of Conditioning Chemotherapy on Lymphocyte Kinetics and Outcomes in LBCL Patients Treated with CAR T-cell Therapy

Paolo Strati 1,2,*, Andrew P Jallouk 1,*, Ryan Sun 3, Jaihee Choi 3, Kaberi Das 1, Hua-Jay Cherng 1, Sairah Ahmed 1, Hun J Lee 1, Swaminathan P Iyer 1, Ranjit Nair 1, Loretta J Nastoupil 1, Raphael E Steiner 1, Chad D Huff 4, Yao Yu 4, Haleigh Mistry 1, Brittany Pulsifer 1, Mansoor Noorani 1, Neeraj Saini 5, Elizabeth J Shpall 5, Partow Kebriaei 5, Christopher R Flowers 1, Jason R Westin 1, Michelle AT Hildebrandt 1,#, Sattva S Neelapu 1,#
PMCID: PMC9904360  NIHMSID: NIHMS1869227  PMID: 36127509

Abstract

Conditioning chemotherapy (CCT) has been shown to be essential for optimal efficacy of chimeric antigen receptor (CAR) T-cell therapy. Here, we determined whether the change in absolute lymphocyte count, referred to as delta lymphocyte index (DLIx), may serve as a surrogate marker for pharmacodynamic effects of CCT and whether it associated with germline genetic variants in patients with large B-cell lymphoma (LBCL).

One-hundred and seventy-one patients were included, of which 86 (50%) received bridging therapy post-leukapheresis. Median DLIx was 0.5 × 109/L (range, 0.01–2.75 X 109/L) and was significantly higher in patients who achieved complete response (p=0.04). On multivariate analysis, low DLIx was associated only with use of bridging therapy (odds ratio 0.4, 95% CI 0.2–0.8, p=0.007). Low DLIx was independently associated with shorter progression-free (p=0.02) and overall survival (p=0.02). DLIx was associated with genetic variations related to drug metabolism and macrophage biology such as ABCB1, MISP and CPVL.

The impact of CCT on lymphocyte count is affected by use of bridging therapy but change in lymphocyte count is independently associated with efficacy. Studies aimed at investigating macrophage biology in this setting may suggest strategies to increase the efficacy of CCT and improve outcomes.

Scientific Category: Immunobiology and Immunotherapy, Lymphoid neoplasia

Introduction

Conditioning chemotherapy (CCT) has been shown to be essential for optimal efficacy of adoptive T-cell therapy including chimeric antigen receptor (CAR) T-cell therapy.1, 2 The favorable impact of CCT is mediated by multiple biological mechanisms, including but not limited to elimination of sinks for homeostatic cytokines (mainly interleukin [IL] 7 and IL-15) and eradication of regulatory T-cells and myeloid-derived suppressor cells.35 The addition of fludarabine to cyclophosphamide has been associated with improved CAR T-cell expansion and persistence as compared to other regimens, and the combination of these agents represents the preferred CCT regimen in patients with relapsed or refractory large B-cell lymphoma (LBCL).5 However, the identification of patients who would benefit from more or less intensive CCT remains challenging. For instance, while patients who receive high-intensity CCT have a greater probability of achieving a favorable cytokine profile, high levels of IL-7 and IL-15 are associated with improved outcome independent of CCT intensity, prompting the question whether pharmacokinetics and pharmacogenomics of fludarabine and cyclophosphamide may play a role.6 Similarly, in clinical trials where the use of CCT was omitted based on a low absolute lymphocyte count (ALC) before CAR T-cell infusion, patients experienced worse outcomes, suggesting that pre-conditioning lymphocyte count alone may not be sufficient to predict the need for CCT.7 In this study, we analyze the impact of CCT on lymphocyte count and assess how its depth of change relates to clinical outcomes, cytokine levels, and genetic variation in patients with LBCL receiving CAR T-cell therapy.

Methods

Methods

Patient selection and CCT regimen

This is a retrospective cohort analysis of 171 consecutive patients with relapsed or refractory LBCL treated with standard of care axicabtagene ciloleucel (axi-cel) at our institution, The University of Texas MD Anderson Cancer Center (MDACC), between 01/2018 and 04/2020. Standard of care was defined as administration of commercial product outside of a clinical trial. Data cut-off was 04/2021. Patients received CCT with cyclophosphamide (500 mg/m2) and fludarabine (30 mg/m2) administered intravenously on days −5, −4 and −3, followed by axi-cel infusion (2 × 106 cells/kg) on day 0. The study was approved by the Institutional Review Board of MD Anderson Cancer Center and conducted in accordance with our institutional guidelines and the principles of the Declaration of Helsinki.

DLIx definition, toxicity and response assessment

Clinical characteristics and laboratory features before CCT (day −5) and on the day of axi-cel infusion (day 0) were collected prospectively. Delta lymphocyte index (DLIx) was defined as the difference in ALC between the day of initiation of CCT and the day of axi-cel infusion. As no association between normalized DLIx (according to baseline ALC) and outcomes was observed, the latter was not used in the final analysis. DLIx was categorized in quartiles: quartile 1 (median 0.19, range 0.01–0.26), quartile 2 (median 0.41, range 0.27–0.52), quartile 3 (median 0.69, range 0.55–0.79), and quartile 4 (median 1.36, range 0.88–2.75). Their association with survival was analyzed, and quartile 1 selected as low (vs high) DLIx based on the strongest association with survival outcomes.

Cytokine release syndrome (CRS) and immune cell associated neurotoxicity syndrome (ICANS) were prospectively graded for up to 30 days after axi-cel infusion, according to the CARTOX grading system from 01/2018 to 04/2019, and according to ASTCT criteria from 05/2019 onward.8, 9 Performance status was defined according to the Eastern Cooperative Oncology Group (ECOG).10 The international prognostic index (IPI) was calculated as previously described.11 Response status was prospectively determined by the Lugano 2014 classification.12

Cytokine measurements and single nucleotide polymorphism analysis

IL-7 and IL-15 were measured by multiplex assays (V-Plex, Meso Scale Diagnostics) in matched plasma samples on day −5 and day 0. Germline DNA for a subset of patients (N=132) was isolated from banked peripheral blood samples using standard procedures. Genotyping was conducted on the Illumina Genome Screening Array (GSAv2.0) with inclusion of 2% replicates for quality control and repeated for samples with call rates less than 95%. Following removal of duplicate samples, imputation was performed to HRC-r1.1 (hg19) using the Michigan Imputation Server.13 Variants with minor allele frequencies <1% were excluded from further analysis. A curated panel of candidate variants were selected from the PharmGKB database based on genetic variants associated with response to cyclophosphamide or fludarabine.14 A total of 144 variants were identified as having at least one significant previous association reported, of which 131 had genotyping data available in the imputed dataset (Supplementary Table 1).

Statistical methods

Association between categorical variables was evaluated using χ2 test or Fisher’s exact test. The difference in a continuous variable between patient groups was evaluated by the Mann-Whitney test. Only factors significant (p-value 0.05) on univariate analysis were included in multivariate models. Wilcoxon signed-rank test was used for cytokine level comparison. Genetic variants were analyzed under a linear additive genetic model with DLIx as the outcome. Additional covariates were included to control for the confounding effects of age, gender, and bridging therapy. Significance of each variant was assessed by one degree of freedom Wald test with the effect allele for the candidate variant analyses based on those reported in PharmGKB, and we first conducted a candidate SNP analysis with 131 total SNPs selected due to their prior associations with pharmacodynamic processes of interest. Additionally, gene-based analyses were conducted using the sequence kernel association test (SKAT), which specifies the genetic effects to be random and then performs a variance components test.15 In further exploratory investigation, a genome-wide single variant analysis was also conducted with the standard Bonferroni-corrected genome-wide association threshold of 5×10−8 used for significance.

Survival outcomes were prospectively collected. Progression-free survival (PFS) was defined as the time from the start of axi-cel infusion to progression of disease, death, or last follow-up (whichever occurred first). Overall survival (OS) was defined as the time from the start of axi-cel infusion to death or last follow-up. PFS and OS were calculated for all patients in the study and for subgroups of patients using Kaplan-Meier estimates and were compared between subgroups using the log-rank test. Multivariable Cox regression analysis was performed to assess the associations between patient characteristics and PFS or OS.

Results

Baseline characteristics and factors associated with low DLIx

One hundred and seventy-one patients were included in the analysis. Baseline characteristics collected before initiation of CCT (day −5) are shown in Table 1. Median ALC at the time of CCT initiation (day −5) was 0.6 X 109/L (range, 0.04–2.8 X 109/L) and below the lower limit of normal (LLN) in 131 (77%) patients. At the time of axi-cel infusion (day 0), median ALC was 0.03 X109/L (range, 0–1.9 X109/L), and median DLIx was 0.5 X 109/L (range, 0.01–2.75 X 109/L)(Figure 1A). Ten (6%) patients experienced a delay between CCT and axi-cel infusion. In these patients, the median time from CCT initiation to axi-cel infusion was 13 days (range, 7–19 days) but no association between delay and DLIx was observed (p=0.79). Eighty-six (50%) patients received bridging therapy: 52 (30%) received chemotherapy, including a platinum-based regimen in 33 patients and a hyper-fractionated cyclophosphamide-based regimen in 19; 13 (8%) received biological therapy, including polatuzumab vedotin with rituximab (no bendamustine) in 9 patients, lenalidomide in 3, ibrutinib in 1 patient; and 21 (12%) received radiation therapy (Table 1). When considering DLIx as a continuous variable, on univariate analysis, the baseline characteristics associated with low DLIx were ALC < LLN (p<0.001), platelet count < LLN (p=0.003) and use of bridging therapy (p=0.02). On multivariate analysis, only the association between low DLIx and use of bridging therapy was maintained (odds ratio [OR] 0.4, 95% confidence interval [CI] 0.2–0.8, p=0.007)(Figure 1B) whereas the association with ALC < LLN was not maintained (Table 1). When considering DLIx as a categorical variable, on univariate analysis, the baseline characteristics associated with low DLIx were IPI score of 3–5 (p=0.003), ALC < LLN (p<0.001), and use of bridging therapy (p=0.02). On multivariate analysis, low DLIx and use of bridging therapy remained independently associated (OR 0.5, 95%CI 0.2–0.9, p=0.04), whereas the association with ALC < LLN was not maintained (Table 2).

Table 1.

Baseline characteristics according to DLIx (as continuous variable)

Total (N=171) Number (%) Median DLIx (109/L) p-uni OR [95%CI] p-multi
DLBCL/HGBCL 133 (78) 0.51 0.76 - -
PMBCL/TFL 38 (22) 0.58
Age > 60 years 78 (46) 0.55 0.72 - -
 <= 60 years 93 (54) 0.51
Male 120 (70) 0.49 0.25 - -
Female 51 (30) 0.62
ECOG PS 2–4 20 (12) 0.32 0.76 - -
 0–1 151 (88) 0.58
Ann Arbor Stage III-IV 140 (82) 0.51 0.91 - -
 Stage I-II 31 (18) 0.55
Extra-nodal sites > 1 109 (64) 0.47 0.20 - -
 0–1 62 (36) 0.63
IPI score 3–5 96 (56) 0.44 0.41 - -
 0–2 75 (44) 0.60
ANC low 35 (20) 0.47 0.52 - -
 normal 136 (80) 0.56
AMC low 11 (6) 0.50 0.95 - -
 normal 160 (94) 0.52
ALC low 131 (77) 0.82 <0.001 0.03 1
 normal 40 (23) 1.4 [0.01–1.1]
Hemoglobin low 163 (95) 0.51 0.68 - -
 normal 8 (5) 0.90
PLT count low 88 (51) 0.40 0.003 0.7 0.27
 high 83 (49) 0.69 [0.3–1.3]
CRP high 113 (66) 0.46 0.11 - -
 normal 58 (34) 0.68
Ferritin high 126 (74) 0.47 0.24 - -
 normal 45 (26) 0.61
LDH high 118 (69) 0.45 0.14 - -
 normal 53 (31) 0.61
eGFR low 31 (18) 0.55 0.91 - -
 high 140 (82) 0.52
Previous therapies > 2 135 (79) 0.48 0.29 - -
 2 36 (21) 0.61
Bridging therapy: yes 86 (50) 0.49 0.02 0.4 0.007
 no 85 (50) 0.60 [0.2–0.8]
Bridging: chemotherapy 52 (30) 0.41 0.18 - -
 other/no 119 (70) 0.57
Refractory disease: yes 134 (78) 0.52 0.90 - -
 no 37 (22) 0.55
Previous auto-SCT: yes 45 (26) 0.55 0.89 - -
 no 126 (74) 0.51
Previous allo-SCT: yes 3 (2) 0.88 0.98 - -
 no 168 (98) 0.55

DLIx, delta lymphocyte index; OR, odds ratio; CI, confidence interval; uni, univariate; multi, multivariate; DLBCL, diffuse large B-cell lymphoma; HGBCL, high grade B-cell lymphoma; PMBCL, primary mediastinal B-cell lymphoma; TFL, transformed follicular lymphoma; ECOG, European Cooperative Oncology Group; PS, performance status; IPI, international prognostic index; ANC, absolute neutrophil count; AMC, absolute monocyte count; ALC, absolute lymphocyte count; PLT, platelet; CRP, C-reactive protein; LDH, lactate dehydrogenase; eGFR, estimated glomerular filtration rate; auto, autologous; allo, allogeneic; SCT, stem cell transplant

Figure 1. Factors associated with DLIx and efficacy outcomes. A) Absolute lymphocyte count on day −5 and day 0. B-D) DLIx in patients with and without bridging therapy (B) and patients with or without complete response after axi-cel therapy (C). The red horizontal bars represent medians for each group. E-F) Progression-free (E) and overall (F) survival in patients with low DLIx (Quartile 1, Q1) and high DLIx (Q2–4).

Figure 1.

The red bar in Figure A and B and C indicates median value with 95% confidence interval

The dotted lines in Figure E and F represent 95% confidence interval

Table 2.

Baseline characteristics according to DLIx (as categorical variable)

Total (N=171) High DLIx
(N=126)
Low DLIx
(N=45)
p-uni OR [95%CI] p-multi
DLBCL/HGBCL 101 (80) 32 (71) 0.22 - -
Age > 60 years 59 (47) 19 (42) 0.61 - -
Male 90 (71) 30 (67) 0.57 - -
ECOG PS 2–4 11 (9) 9 (20) 0.06 - -
Ann Arbor Stage III-IV 102 (81) 38 (84) 0.66 - -
Extra-nodal sites > 1 75 (60) 34 (76) 0.07 - -
IPI score 3–5 62 (49) 34 (76) 0.003 0.7 [0.2–1.8] 0.42
ANC low 23 (18) 12 (27) 0.28 - -
AMC low 6 (5) 5 (11) 0.16 - -
ALC low 86 (68) 45 (100) <0.001 0.03 [0.01–1.1] 1
Hemoglobin low 119 (94) 44 (98) 0.68 - -
PLT count low 59 (47) 29 (64) 0.06 - -
CRP high 80 (63) 33 (73) 0.27 - -
Ferritin high 91 (72) 35 (78) 0.56 - -
LDH high 78 (62) 40 (89) 0.10 - -
eGFR low 22 (17) 9 (20) 0.82 - -
Previous therapies > 2 98 (78) 37 (82) 0.67 - -
Bridging therapy: yes 56 (44) 30 (67) 0.02 0.5 [0.2–0.9] 0.04
Bridging: chemotherapy 35 (28) 17 (38) 0.26 - -
Refractory disease: yes 98 (78) 36 (80) 0.84 - -
Previous auto-SCT: yes 38 (30) 7 (16) 0.08 - -
Previous allo-SCT: yes 3 (2) 0 (0) 0.57 - -

DLIx, delta lymphocyte index; OR, odds ratio; CI, confidence interval; uni, univariate; multi, multivariate; DLBCL, diffuse large B-cell lymphoma; HGBCL, high grade B-cell lymphoma; PMBCL, primary mediastinal B-cell lymphoma; TFL, transformed follicular lymphoma; ECOG, European Cooperative Oncology Group; PS, performance status; IPI, international prognostic index; ANC, absolute neutrophil count; AMC, absolute monocyte count; ALC, absolute lymphocyte count; PLT, platelet; CRP, C-reactive protein; LDH, lactate dehydrogenase; eGFR, estimated glomerular filtration rate; auto, autologous; allo, allogeneic; SCT, stem cell transplant

Association of DLIx with toxicity and efficacy outcomes

Overall, 13 (8%) patients had grade (G) ≥3 CRS, and 61 (36%) had G≥3 ICANS. There was no association between DLIx and incidence of G≥3 CRS (p=0.28) or G≥3 ICANS (p=0.43). Of 164 patients evaluable for response, complete responses (CR) was achieved in 89 (54%) patients. DLIx was significantly higher in patients who achieved CR as compared to those who did not (p=0.04), and CR rate of 43% in patients with low DLIx and 58% in patients with high DLIx (defined as quartile 1 vs. quartile 2–4)(p=0.09)(Figure 1CD).

After a median follow-up of 26 months (95% CI, 24–28 months), 116 (68%) patients progressed and/or died, and median progression-free survival (PFS) was 6 months (95% CI, 4–8 months). On univariate analysis, baseline characteristics associated with shorter median PFS were ECOG performance status 2–4 (3 vs 7 months, p=0.02), IPI score 3–5 (3 vs 16 months, p<0.001), use of bridging therapy (4 vs 8 months, p=0.02) and low DLIx (3 vs 9 months, p=0.02)(Figure 1E); on multivariate analysis, low DLIx remained independently associated with shorter median PFS (hazard ratio [HR] 0.6, 95%CI 0.4–0.9; p=0.04)(Table 3). Of interest, the association between low DLIx and shorter median PFS was observed also when limiting the analysis to the 85 patients who did not receive bridging therapy (2 vs 26 months, p=0.03).

Table 3.

Univariate and multivariate analysis for progression-free and overall survival

Total (N=171) Events Median PFS (months) p-uni HR [95%CI] p-multi
DLBCL/HGBCL 75 6 0.59 - -
PMBCL/TFL 19 7
Age > 60 years 43 6 0.91 - -
 <= 60 years 51 5
Male 68 5 0.42 - -
Female 26 8
ECOG PS 2–4 15 3 0.02 0.8 [0.4–1.4] 0.36
 0–1 79 7
IPI score 3–5 64 3 <0.001 0.5 [0.3–0.8] 0.002
 0–2 30 16
Previous therapies > 2 78 5 0.11 - -
 2 16 14
Bridging therapy: yes 52 4 0.02 0.7 [0.5–1.1] 0.16
 no 42 8
Refractory disease: yes 79 5 0.08 - -
 no 15 26
DLIx: low 35 3 0.02 0.6 [0.4–0.9] 0.04
 high 81 9
Total (N=171) Events Median OS (months) p-uni HR [95%CI] p-multi
DLBCL/HGBCL 45 16 0.63 - -
PMBCL/TFL 11 18
Age > 60 years 29 16 0.30 - -
 <= 60 years 27 18
Male 39 17 0.77 - -
Female 17 17
ECOG PS 2–4 10 8 0.03 0.8 [0.4–1.5] 0.43
 0–1 46 18
IPI score 3–5 44 14 <0.001 0.3 [0.2–0.6] 0.01
 0–2 12 22
Previous therapies > 2 47 17 0.19 - -
 2 9 17
Bridging therapy: yes 30 15 0.20 - -
 no 36 18
Refractory disease: yes 49 16 0.07 - -
 no 7 27
DLIx: low 29 7 0.02 0.5 [0.3–0.9] 0.02
 high 65 26

DLIx, delta lymphocyte index; HR, hazard ratio; CI, confidence interval; uni, univariate; multi, multivariate; DLBCL, diffuse large B-cell lymphoma; HGBCL, high grade B-cell lymphoma; PMBCL, primary mediastinal B-cell lymphoma; TFL, transformed follicular lymphoma; ECOG, European Cooperative Oncology Group; PS, performance status; IPI, international prognostic index

At most recent follow-up, 94 (55%) patients had died and median OS was 17 months (95% CI, 10–24 months). On univariate analysis, baseline characteristics associated with shorter median OS were ECOG performance status 2–4 (8 vs 18 months, p=0.03), IPI score 3–5 (14 vs 22 months, p<0.001) and low DLIx (7 vs 26 months, p=0.02)(Figure 1F); on multivariate analysis, low DLIx remained independently associated with shorter median OS (HR 0.5, 95%CI 0.3–0.9; p=0.02)(Table 3). Of interest, the association between low DLIx and shorter median OS was observed also when limiting the analysis to the 85 patients who did not receive bridging therapy (5 months vs not reached, p<0.001).

DLIx and cytokine homeostasis

Matched plasma samples collected on day −5 and day 0 were available for 40 patients, including 8 patients with low DLIx and 32 patients with high DLIx. Mean plasma IL-7 levels were 3.65 pg/mL (range, 0.16–15.91) on day −5, and 15.47 pg/mL (range, 0.38–58.55) on day 0, with a mean increase of 11.8 pg/mL (range, 0.31–54.45). Mean plasma IL-15 levels were 5.07 pg/mL (range, 0.93–34.66) on day −5, and 25.64 pg/mL (range, 1.85–66.61) on day 0, with a mean increase of 20.57 pg/mL (range, 0.46–62.75).When comparing patients with low DLIx to those with high DLIx, there were no significant differences in mean plasma IL-7 and IL-15 levels on day −5 (p=0.28 and p=0.16, respectively), day 0 (p=0.16 and 0.11, respectively)(Figure 2AB). No significant difference in the delta change in IL-7 and IL-15 levels between these days was observed when comparing the 2 groups (p=0.43 and p=0.17, respectively)(Figures 2C). Similarly, no significant associations were observed for IL-2 (data not shown).

Figure 2.

Figure 2.

Change in plasma levels of IL-7 and IL-15 between day −5 and day 0 according to DLIx. A. Plasma IL-7 levels. B. Plasma IL-15 levels. C. Delta change in plasma IL-7 and IL-15 levels

Germline genetic variation and DLIx

At the conservative Bonferroni p-value threshold of 0.05 / 131 = 0.00038, none of the candidate variants previously implicated in pharmacogenetic events related to cyclophosphamide or fludarabine exposure were significantly associated with DLIx (Supplemental Table 1). However, one variant (rs8110536) in MISP was nearing Bonferroni significance with a p-value of 0.00063, and the G allele was associated with a high DLIx (β: 0.26, SE: 0.074)(Table 4). In gene-based analysis, MISP was significantly associated with DLIx (p=0.0059), suggesting that multiple genetic variants across this gene may be important in mediating the DLIx phenotype rather than the effect being driven by a single variant (Figure 3A). Of interest, within the top 12 variants with p<0.05 in the candidate variant analysis, 5 were within ABCB1, and the gene-based p-value for ABCB1 was significant (p=0.014). In the exploratory genome-wide analysis, a region on chromosome 7 contained several variants with individual p-values in the range of 10−7 (Figure 3B). While its significance was below the threshold required for genome-wide analyses, this region encompassed CPVL, which was significant in a gene-based analysis with a p-value of 3.70×10−5.

Table 4.

Candidate Pharmacogenomic Variants Associated with DLIx Phenotype

Variant Gene Chr. Effect Allele *β (SE) p-value
rs8110536 MISP 19 G 0.26 (0.074) 0.00063
rs1128503 ABCB1 7 G 0.18 (0.057) 0.0025
rs10276036 ABCB1 7 T 0.18 (0.057) 0.0025
rs1801280 NAT2 8 C −0.17 (0.058) 0.0045
rs2032582 ABCB1 7 A −0.15 (0.056) 0.0099
rs1045642 ABCB1 7 G 0.13 (0.053) 0.015
rs4148737 ABCB1 7 C 0.14 (0.061) 0.021
rs2070744 NOS3 7 C 0.14 (0.061) 0.028
rs738409 PNPLA3 22 G −0.14 (0.066) 0.035
rs246221 ABCC1 16 C 0.12 (0.057) 0.036
rs639174 DROSHA 5 T 0.14 (0.066) 0.040
rs17222723 ABCC2 10 A 0.23 (0.12) 0.045

Effect allele based on previous reports in PharmGKB (Supplemental Table 1)

*

linear additive genetic model with change in absolute lymphocyte count as the outcome with age at diagnosis, gender, and bridging therapy included in model

Figure 3. Association between germline genetic variation and DLIx. A) Association between genetic variants of MISP and DLIx. B) Association between genetic variants of CPVL and DLIx.

Figure 3.

Dotted line represents the Bonferroni p-value threshold for the candidate gene analysis (A) and genome-wide analysis (B).

Discussion

In this study, we demonstrate that the impact of CCT on lymphocyte count is affected by use of bridging therapy but change in lymphocyte count is indipendently associated with efficacy after CAR T-cell therapy in LBCL. Bridging therapy is used in about one half of individuals in real-world practice. These patients typically have higher-risk disease and exemplify the need to identify optimal bridging strategies.16, 17 The pattern of utilization of bridging therapies remains very heterogeneous, and no preferable strategy has been identified to date.18, 19 The small number of patients treated with bridging radiation in our study did not allow us to assess its specific impact on DLIx. However, radiation therapy has been shown to be a safe and effective bridging approach for patients with LBCL, not only reducing myelosuppression compared to bridging chemotherapy, but also potentially favorably impacting the tumor immune microenvironment.2022 A more detailed understanding of the biological impact of radiation therapy and other chemotherapy-free bridging strategies would aid in the selection of optimal bridging therapy and could significantly impact CAR T-cell efficacy.

In our analysis, despite confirming an increase in plasma IL-7 and IL-15 levels as effect of CCT both among patients with low and high DLIx, the difference was not significantly different according to DLIx. While an increase in the levels of these cytokines has been associated with clinical response after anti-CD19 CAR T-cell therapy in previous studies,4, 6 our findings suggest that this observation may not be solely related to the decrease in lymphocyte count following CCT, and other biological mechanisms may be responsible for the prognostic effect of DLIx.

To this regard, we observed an association between DLIx and genetic variants involving three genes. The first gene was ABCB1, which encodes for P-glycoprotein 1 (also known as multidrug resistance protein 1), a well-known membrane efflux pump involved in chemotherapy metabolism.23 This finding likely explains the variability in DLIx across different individuals but does not provide insight into the biological mechanisms responsible for the association of DLIx with CAR T-cell efficacy, though ABCB1 is also variably expressed in T cells.24 Of note, no significant association was observed in our study with genotypic variants of DCK, coding for deoxycytidine kinase, or of CYP2C19 and CYP2B6, coding for cytochrome P450 family members, all of which are also involved in the metabolism of fludarabine and cyclophosphamide.2527

The other two genes whose variants were associated with DLIx included MISP, coding for mitotic spindle positioning protein, and CPVL, coding for carboxypeptidase vitellogenic like protein. Interestingly, the former is an F-actin bundling protein involved in actin-based cytoskeletal reorganizations, which are crucial for macrophage proliferation, phagocytosis and polarization.28, 29 The latter is a serine carboxypeptidase with exclusive expression in macrophages.30, 31 These findings raise the possibility that macrophages may play a role in mediating the effect of CCT. Indeed, increases in serum monocyte chemoattractant protein-1 (MCP-1) concentration in response to lymphodepletion have been associated with increased probability of achieving a CR and improved PFS in patients with LBCL treated with axi-cel.6 Eradication of myeloid-derived suppressor cells (MDSCs) is also thought to be an important function of CCT.1, 6 Of note, transcriptomic analysis performed on LBCL samples before axi-cel infusion have demonstrated that macrophage gene signatures are enriched in patients who relapse early.32 Furthermore, high levels of circulating monocytic MDSCs have been associated with lower peak CAR T-cell expansion.32, 33 Several agents which impact macrophage function and induce an anti-tumoral phenotype have been shown to be safe and effective for the treatment of patients with LBCL.3438 Investigation of these agents either as bridging therapy or in combination with CCT need to be explored to determine whether they could lead to new strategies to improve the efficacy of CAR T-cell therapy.

We acknowledge multiple limitations of this study, including its small sample size, its retrospective and single-center nature (and hence the need of a prospective validation), the change in toxicity grading systems over time, and the lack of robust power for the cytokine analysis. While these features may limit the general applicability of our findings, the DLIx is easily calculated using basic clinical data and could be readily assessed in other settings. Our observation regarding the impact of bridging therapy is also consistent with those of previous studies.16 Furthermore, even with only 132 patients in our study, both MISP and CPVL were significant in gene-based analyses and nearly reaching the stringent Bonferroni threshold for significance in individual variant analyses, suggesting that these genes are highly likely influence the response to CCT.

In conclusion, the impact of CCT on lymphocyte count is associated with efficacy after CAR T-cell therapy and is affected by use of bridging therapy. Studies aimed at investigating macrophage biology in this setting may suggest strategies to increase the efficacy of CCT and improve outcomes for patients with LBCL.

Supplementary Material

Supplementary Table 1

Key Points.

  • DLIx after CCT is lower in patients who receive bridging therapy and is associated with clinical outcomes after CAR T-cell therapy.

  • Germline genetic variants associated with macrophage biology affect DLIx and suggest that macrophages may play a role in response to CCT.

Acknowledgments

This research is supported in part by the University of Texas MD Anderson Cancer Center B-cell Lymphoma Moonshot (SSN) and The University of Texas MD Anderson Cancer Center Support Grant from National Institutes of Health (P30 CA016672) and by the Shirley Stein Scientific Endowment Research Award.

PS salary is supported by the Lymphoma Research Foundation Career Development Award, the Leukemia Lymphoma Society Career Development Program, the Kite Gilead Scholar in Clinical Research Award, the Sabin Fellowship Award and by the R21 NIH grant. The MD Anderson Lymphoma Tissue Bank is supported by the KW Cares Foundation.

Disclosure of Conflict of Interest

PS is a consultant or has served on advisory boards for Kite Gilead, Sobi, Roche-Genentech, Hutchinson MediPharma, ADC Therapeutics, Incyte Morphosys and TG Therapeutics, and received research funds from Sobi, Astrazeneca-Acerta, ADC Therapeutics and ALX Oncology. S.S.N. has received personal fees from Kite, a Gilead Company, Merck, Bristol Myers Squibb, Novartis, Celgene, Pfizer, Allogene Therapeutics, Sellas Life Sciences, Cell Medica/Kuur/Athenex, Incyte, Precision Biosciences, Legend Biotech, Adicet Bio, Calibr, Unum Therapeutics, Bluebird Bio, and Sana Biotechnology; research support from Kite, a Gilead Company, Bristol Myers Squibb, Merck, Poseida, Cellectis, Celgene, Karus Therapeutics, Unum Therapeutics, Allogene Therapeutics, Precision Biosciences, Acerta, and Adicet Bio; royalties from Takeda Pharmaceuticals; and has intellectual property related to cell therapy.

Data Availability Statement

The datasets generated during and/or analysed during the current study are available from the corresponding author on reasonable request.

References

  • 1.Muranski P, Boni A, Wrzesinski C, Citrin DE, Rosenberg SA, Childs R, et al. Increased intensity lymphodepletion and adoptive immunotherapy--how far can we go? Nat Clin Pract Oncol 2006. Dec; 3(12): 668–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Neelapu SS. CAR-T efficacy: is conditioning the key? Blood 2019. Apr 25; 133(17): 1799–1800. [DOI] [PubMed] [Google Scholar]
  • 3.Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Palmer DC, Spiess PJ, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 2005. Oct 3; 202(7): 907–912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, et al. Lymphoma Remissions Caused by Anti-CD19 Chimeric Antigen Receptor T Cells Are Associated With High Serum Interleukin-15 Levels. J Clin Oncol 2017. Jun 1; 35(16): 1803–1813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest 2016. Jun 1; 126(6): 2123–2138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hirayama AV, Gauthier J, Hay KA, Voutsinas JM, Wu Q, Gooley T, et al. The response to lymphodepletion impacts PFS in patients with aggressive non-Hodgkin lymphoma treated with CD19 CAR T cells. Blood 2019. Apr 25; 133(17): 1876–1887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Andreadis C, Tam CS, Borchmann P, Jaeger U, McGuirk JP, Holte H, et al. Correlation of Bridging and Lymphodepleting Chemotherapy with Clinical Outcomes in Patients with Relapsed/Refractory Diffuse Large B-Cell Lymphoma Treated with Tisagenlecleucel. Blood 2019; 134(Supplement_1): 2883–2883. [Google Scholar]
  • 8.Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol 2018. Jan; 15(1): 47–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lee DW, Santomasso BD, Locke FL, Ghobadi A, Turtle CJ, Brudno JN, et al. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biol Blood Marrow Transplant 2019. Apr; 25(4): 625–638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET, et al. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol 1982. Dec; 5(6): 649–655. [PubMed] [Google Scholar]
  • 11.Sehn LH, Berry B, Chhanabhai M, Fitzgerald C, Gill K, Hoskins P, et al. The revised International Prognostic Index (R-IPI) is a better predictor of outcome than the standard IPI for patients with diffuse large B-cell lymphoma treated with R-CHOP. Blood 2007. Mar 1; 109(5): 1857–1861. [DOI] [PubMed] [Google Scholar]
  • 12.Cheson BD, Fisher RI, Barrington SF, Cavalli F, Schwartz LH, Zucca E, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol 2014. Sep 20; 32(27): 3059–3068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Das S, Forer L, Schonherr S, Sidore C, Locke AE, Kwong A, et al. Next-generation genotype imputation service and methods. Nat Genet 2016. Oct; 48(10): 1284–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Whirl-Carrillo M, Huddart R, Gong L, Sangkuhl K, Thorn CF, Whaley R, et al. An Evidence-Based Framework for Evaluating Pharmacogenomics Knowledge for Personalized Medicine. Clin Pharmacol Ther 2021. Sep; 110(3): 563–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wu MC, Lee S, Cai T, Li Y, Boehnke M, Lin X. Rare-variant association testing for sequencing data with the sequence kernel association test. Am J Hum Genet 2011. Jul 15; 89(1): 82–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Nastoupil LJ, Jain MD, Feng L, Spiegel JY, Ghobadi A, Lin Y, 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. Sep 20; 38(27): 3119–3128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Jacobson CA, Hunter BD, Redd R, Rodig SJ, Chen PH, Wright K, et al. Axicabtagene Ciloleucel in the Non-Trial Setting: Outcomes and Correlates of Response, Resistance, and Toxicity. J Clin Oncol 2020. Sep 20; 38(27): 3095–3106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Dwivedy Nasta S, Hughes ME, Namoglu EC, Landsburg DJ, Chong EA, Barta SK, et al. A Characterization of Bridging Therapies Leading up to Commercial CAR T-Cell Therapy. Blood 2019; 134(Supplement_1): 4108–4108. [Google Scholar]
  • 19.Paillassa J, Vercellino L, Di Blasi R, Bernard S, Moatti H, Bommier C, et al. Impact of Bridging Chemotherapy on Clinical Outcomes of CD19 CAR T Therapy in Relapse/Refractory Diffuse Large B- Cell Lymphoma in Real World Experience. Blood 2019; 134(Supplement_1): 2886–2886. [Google Scholar]
  • 20.Sim AJ, Jain MD, Figura NB, Chavez JC, Shah BD, Khimani F, et al. Radiation Therapy as a Bridging Strategy for CAR T Cell Therapy With Axicabtagene Ciloleucel in Diffuse Large B-Cell Lymphoma. Int J Radiat Oncol Biol Phys 2019. Dec 1; 105(5): 1012–1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Imber B, Palomba ML, DeSelm C, Batlevi CL, Dahi PB, Giralt SA, et al. MSKCC Early Experience Using Radiotherapy As a Bridging Strategy for Relapsed Diffuse Large B Cell Lymphoma before CD19 CAR T Therapy. Blood 2019; 134(Supplement_1): 3238–3238. [Google Scholar]
  • 22.Pinnix CC, Gunther JR, Dabaja BS, Strati P, Fang P, Hawkins MC, et al. Bridging therapy prior to axicabtagene ciloleucel for relapsed/refractory large B-cell lymphoma. Blood Adv 2020. Jul 14; 4(13): 2871–2883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bertholee D, Maring JG, van Kuilenburg AB. Genotypes Affecting the Pharmacokinetics of Anticancer Drugs. Clin Pharmacokinet 2017. Apr; 56(4): 317–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dimeloe S, Frick C, Fischer M, Gubser PM, Razik L, Bantug GR, et al. Human regulatory T cells lack the cyclophosphamide-extruding transporter ABCB1 and are more susceptible to cyclophosphamide-induced apoptosis. Eur J Immunol 2014. Dec; 44(12): 3614–3620. [DOI] [PubMed] [Google Scholar]
  • 25.Rivero A, Rapado I, Tomas JF, Montalban C, de Ona R, Paz-Carreira J, et al. Relationship between deoxycytidine kinase (DCK) genotypic variants and fludarabine toxicity in patients with follicular lymphoma. Leuk Res 2011. Apr; 35(4): 431–437. [DOI] [PubMed] [Google Scholar]
  • 26.Johnson GG, Lin K, Cox TF, Oates M, Sibson DR, Eccles R, et al. CYP2B6*6 is an independent determinant of inferior response to fludarabine plus cyclophosphamide in chronic lymphocytic leukemia. Blood 2013. Dec 19; 122(26): 4253–4258. [DOI] [PubMed] [Google Scholar]
  • 27.Ekhart C, Rodenhuis S, Smits PH, Beijnen JH, Huitema AD. Relations between polymorphisms in drug-metabolising enzymes and toxicity of chemotherapy with cyclophosphamide, thiotepa and carboplatin. Pharmacogenet Genomics 2008. Nov; 18(11): 1009–1015. [DOI] [PubMed] [Google Scholar]
  • 28.Maier B, Kirsch M, Anderhub S, Zentgraf H, Kramer A. The novel actin/focal adhesion-associated protein MISP is involved in mitotic spindle positioning in human cells. Cell Cycle 2013. May 1; 12(9): 1457–1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kumeta M, Gilmore JL, Umeshima H, Ishikawa M, Kitajiri S, Horigome T, et al. Caprice/MISP is a novel F-actin bundling protein critical for actin-based cytoskeletal reorganizations. Genes Cells 2014. Apr; 19(4): 338–349. [DOI] [PubMed] [Google Scholar]
  • 30.Mahoney JA, Ntolosi B, DaSilva RP, Gordon S, McKnight AJ. Cloning and characterization of CPVL, a novel serine carboxypeptidase, from human macrophages. Genomics 2001. Mar 15; 72(3): 243–251. [DOI] [PubMed] [Google Scholar]
  • 31.Harris J, Schwinn N, Mahoney JA, Lin HH, Shaw M, Howard CJ, et al. A vitellogenic-like carboxypeptidase expressed by human macrophages is localized in endoplasmic reticulum and membrane ruffles. Int J Exp Pathol 2006. Feb; 87(1): 29–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jain MD, Zhao H, Wang X, Atkins R, Menges M, Reid K, et al. Tumor interferon signaling and suppressive myeloid cells are associated with CAR T-cell failure in large B-cell lymphoma. Blood 2021. May 13; 137(19): 2621–2633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Strati P, Neelapu SS. CAR-T failure: beyond antigen loss and T cells. Blood 2021. May 13; 137(19): 2567–2568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ansell SM, Flinn IW, Maris MB, O’Connor OA, Lesokhin A, Advani AS, et al. TTI-621 (SIRPaFc), an Immune Checkpoint Inhibitor Blocking the CD47 “Do Not Eat” Signal, Induces Objective Responses in Patients with Advanced, Relapsed/Refractory Diffuse Large B-Cell Lymphoma (DLBCL). Blood 2017. Dec 7; 130. [Google Scholar]
  • 35.Advani RH F I, Popplewell L, Forero-Torres A, Bartlett NL, Ghosh N, Kline JP, et al. Activity and tolerabilty of the first-in-class anti-CD47 antibody Hu5F9-G4 with rituximab tolerated in relapsed/refractory non-Hodgkin lymphoma: Initial phase 1b/2 results. J Clin Oncol 2018; 36(suppl; abstr 7504). [Google Scholar]
  • 36.Lakhani NJ L P, Hafez N, Krishnamurthy A, O’Rourke TJ, Kamdar MK, Fanning P, et al. A phase 1 study of ALX148, a CD47 blocker, alone and in combination with established anticancer antibodies in patients with advanced malignancy and non-Hodgkin lymphoma. J Clin Oncol 2018; 36(suppl; abstr 3068). [Google Scholar]
  • 37.Kim TM, Lakhani N, Gainor J, Kamdar M, Fanning P, Squifflet P, et al. A Phase 1 Study of ALX148, a CD47 Blocker, in Combination with Rituximab in Patients with Non-Hodgkin Lymphoma. Blood 2019; 134(Supplement_1): 1953–1953. [Google Scholar]
  • 38.Advani R, Flinn I, Popplewell L, Forero A, Bartlett NL, Ghosh N, et al. CD47 Blockade by Hu5F9-G4 and Rituximab in Non-Hodgkin’s Lymphoma. N Engl J Med 2018. Nov 1; 379(18): 1711–1721. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Supplementary Table 1

Data Availability Statement

The datasets generated during and/or analysed during the current study are available from the corresponding author on reasonable request.

RESOURCES