• Axicel was independently associated with higher ICANS and CRS severity and higher efficacy than investigational JCAR014.

  • Higher preleukapheresis LDH, largest lesion diameter, and lower ALC were independently associated with a lower efficacy.

CD19-targeted chimeric antigen receptor-engineered (CD19 CAR) T cells are novel therapies showing great promise for patients with relapsed or refractory (R/R) aggressive B-cell non-Hodgkin lymphoma (B-NHL). Single-arm studies showed significant variations in outcomes across distinct CD19 CAR T-cell products. To estimate the independent impact of the CAR T-cell product type on outcomes, we retrospectively analyzed data from 129 patients with R/R aggressive B-NHL treated with cyclophosphamide and fludarabine lymphodepletion followed by either a commercially available CD19 CAR T-cell therapy (axicabtagene ciloleucel [axicel] or tisagenlecleucel [tisacel]), or the investigational product JCAR014 on a phase 1/2 clinical trial (NCT01865617). After adjustment for age, hematopoietic cell transplantation-specific comorbidity index, lactate dehydrogenase (LDH), largest lesion diameter, and absolute lymphocyte count (ALC), CAR T-cell product type remained associated with outcomes in multivariable models. JCAR014 was independently associated with lower cytokine release syndrome (CRS) severity compared with axicel (adjusted odds ratio [aOR], 0.19; 95% confidence interval [CI]; 0.08-0.46), with a trend toward lower CRS severity with tisacel compared with axicel (aOR, 0.47; 95% CI, 0.21-1.06; P = .07). Tisacel (aOR, 0.17; 95% CI, 0.06-0.48) and JCAR014 (aOR, 0.17; 95% CI, 0.06-0.47) were both associated with lower immune effector cell-associated neurotoxicity syndrome severity compared with axicel. Lower odds of complete response (CR) were predicted with tisacel and JCAR014 compared with axicel. Although sensitivity analyses using either positron emission tomography- or computed tomography-based response criteria also suggested higher efficacy of axicel over JCAR014, the impact of tisacel vs axicel became undetermined. Higher preleukapheresis LDH, largest lesion diameter, and lower ALC were independently associated with lower odds of CR. We conclude that CD19 CAR T-cell product type independently impacts toxicity and efficacy in R/R aggressive B-NHL patients.

CD19-targeted chimeric antigen receptor-engineered (CD19 CAR) T cells are novel therapies showing great promise for patients with relapsed or refractory (R/R) aggressive B-cell non-Hodgkin lymphoma (B-NHL). To date, 4 CD19 CAR T-cell products for R/R aggressive B-NHL are already US Food and Drug Administration (FDA)-approved and commercially available: axicabtagene ciloleucel (axicel), tisagenlecleucel (tisacel), brexucabtagene autoleucel, and lisocabtagene maraleucel. Overall, clinical trial1-6 and “real-world”7,8 data showed promising efficacy in R/R B-NHL patients across all CAR T-cell products compared with historical reports,9 with overall response rates and complete response (CR) rates ranging from 51% to 93% and 40% to 64%, respectively. Tempering their high efficacy, CD19 CAR T-cell therapy can be associated with severe, potentially life-threatening toxicities,10-12 such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Published data suggest important variations in the toxicity and efficacy rates across CAR T-cell products, but these studies were limited by the absence of comparative groups and did not specifically adjust estimates for key confounders. Using retrospective data from 129 R/R aggressive B-NHL patients treated with the FDA-approved CAR T-cell products axicel or tisacel, or the investigational product JCAR014, we aimed to estimate the independent impact of the CAR T-cell product type on toxicity and efficacy.

Study design and patient selection

We performed a retrospective analysis of R/R aggressive B-NHL patients treated at our institutions with cyclophosphamide and fludarabine lymphodepletion followed by the infusion of CD19 CAR T cells. Tisacel and axicel patients were treated per standard of care using FDA-approved, commercially available products. The median tisacel dose was 2.2 × 108 CAR T cells (interquartile range [IQR], 1.4-2.85; range, 0.31-4). The exact axicel dose was not measured; the target axicel dose per the FDA label was 2 × 106 CAR-positive viable T cells/kg, with a maximum of 2 × 108 CAR-positive viable T cells. Investigational, defined CD4+/CD8+-composition CD19 CAR T-cells (JCAR014) were administered in all patients at the dose of 2 × 106/kg on a phase 1/2 clinical trial (NCT01865617) as previously described.4,5 All JCAR014 patients included in our dataset were enrolled and apheresed between March 2015 and January 2017, before the approval by the FDA of axicel (October 2017) and tisacel (May 2018) for patients with R/R large B-cell lymphoma. Twenty-nine of 30 JCAR014 patients were infused before FDA approval of axicel and tisacel. One JCAR014 patient was apheresed in November 2016 and infused in February 2018. Details regarding inclusion and exclusion criteria for screening and JCAR014 treatment are detailed in the supplemental Material, available on the Blood Web site. History of prior transplant and bridging therapy were allowed in the study, and there was no absolute lymphocyte count (ALC)-based or T-cell function-based criteria to limit study inclusion. Since we had previously demonstrated a significant impact of the CAR T-cell dose and cyclophosphamide and fludarabine lymphodepletion on outcomes,5,13 JCAR014 patients treated with a CAR T-cell dose of 2 × 105 or 2 × 107/kg and JCAR014 patients who received lymphodepletion other than cyclophosphamide and fludarabine were excluded from our analysis, as detailed in the study flowchart (Figure 1). This study was conducted with the approval of the Fred Hutchinson Cancer Research Center Institutional Review Board and in accordance with the Declaration of Helsinki.

Figure 1.

Patient selection. FHCRC, Fred Hutchinson Cancer Research Center; JCAR014, investigational CD8+:CD4+ 1:1 defined-composition CD19-targeted CAR T cells; OHSU, Oregon Health and Science University. *Dose dense cohort, planned second infusion of CAR T cells 10-21 days after the first infusion without additional lymphodepletion.

Figure 1.

Patient selection. FHCRC, Fred Hutchinson Cancer Research Center; JCAR014, investigational CD8+:CD4+ 1:1 defined-composition CD19-targeted CAR T cells; OHSU, Oregon Health and Science University. *Dose dense cohort, planned second infusion of CAR T cells 10-21 days after the first infusion without additional lymphodepletion.

Close modal

Toxicity assessment and management

In axicel and tisacel patients, peak CRS and ICANS severity after CD19 CAR T-cell therapy were graded retrospectively using chart review according to the ASTCT (American Society of Transplant and Cellular Therapy) consensus grading system14 and the CTCAE (Common Terminology Criteria for Adverse Events) 4.03 criteria, respectively. In patients receiving investigational JCAR014, peak CRS and ICANS severity were initially graded prospectively per protocol using the CTCAE 4.03 criteria; peak CRS severity was then retrospectively regraded using the 2021 ASTCT criteria to apply the same criteria across all product types. CRS and ICANS clinical management were conducted according to the Yescarta15 and Kymriah16 Risk Evaluation and Mitigation Strategy programs and according to established institutional guidelines. The protocol-recommended CRS and ICANS management for JCAR014 patients is shown in the supplemental Material. Severe cytopenia and infections were defined as grade ≥3 per CTCAE 4.03 criteria.

Response assessment

Response to CAR T-cell therapy was defined as the best response within 3 months after CAR T-cell infusion using Lugano 2014 positron emission tomography-computed tomography (PET-CT)–based response criteria or CT-based response criteria17 when a PET was not performed.

Statistical analyses

Comparisons between groups of continuous variables were made using the Kruskal Wallis test and of categorical variables using Pearson's χ-squared test or Fisher’s exact test. All P values are 2-sided and were not corrected for multiplicity. We hypothesized several preleukapheresis variables would confound the effect of the CAR T-cell product type on both toxicity and efficacy. Based on past literature and subject-matter knowledge, we prespecified variables suspected to impact both the choice of the CAR T-cell product and outcomes and/or known to independently impact outcomes. The hypothesized causal relationships between variables were built into a directed diacyclic graph (DAG) (supplemental Figure 1). DAGs are an established framework to address confounding using observational nonrandomized data.18-21 Using the R package Dagitty22 to analyze our DAG, the following predictors were selected as the minimal adjustment set of covariates to estimate the direct effect of the CAR T-cell product type on outcomes: CAR T-cell product type, lactate dehydrogenase (LDH), largest lesion diameter, age, and hematopoietic cell transplantation-specific comorbidity index (HCT-CI). While not considered a confounder, we also included the preleukapheresis ALC. Next, we applied multivariable proportional odds logistic regression to model peak CRS (grade 0, grade 1, grade 2-4) and ICANS grade (grade 0, grade 1, grade 2, grade 3-4), and logistic regression to model day-28 response (complete or partial response vs stable or progressive disease) including the predictors in the minimal sufficient adjustment set, as described above. Missing preleukapheresis largest lesion diameter data (n = 24) were imputed using predictive mean matching with 10 imputations, using the rms::aregImpute R function. Multivariable models were fitted to all imputed datasets using the rms::fit.mult.impute() R function. For continuous variables, all reported odds ratios were rescaled to reflect an increase from the first (25% percentile) to the third quartile (75% percentile), unless stated otherwise. We used the rms::calibrate R function for internal validation of model calibration, which generates bias-corrected smooth calibration curves using bootstrapping (200 repetitions). All statistical analyses were performed using RStudio software (version 1.2.5033, RStudio, Boston, MA). The list of R packages used is available at the end of the supplemental Material. A Reproducible Data Supplement, including the R code to replicate our analyses and including tables, figures, model outputs, analysis of covariance tables, and calibration curves, is available at https://drjgauthier.github.io/lbclcartcomparison/. Datasets are available upon request to the corresponding author.

Patient, disease, and treatment characteristics across CAR T-cell products

Axicel, tisacel, and JCAR014 were administered in 68 (53%), 31 (24%), and 30 (23%) patients, respectively. Most patient and disease characteristics before leukapheresis were comparable across the 3 CAR T-cell products; specifically, we could not demonstrate statistically significant differences in HCT-CI, LDH, Ann Arbor stage, largest lesion diameter, or extranodal disease (Table 1). We noted a higher proportion of patients with non-DLBCL large B-cell histologies in the JCAR014 group (23%) compared with axicel (6%) and tisacel (3%). Lymphoma histologies for JCAR014 patients are detailed in supplemental Table 1. We observed a higher percentage of patients receiving bridging chemotherapy after leukapheresis in those treated with axicel and tisacel than JCAR014 (P < .001). The number of prior therapies was higher in JCAR014 patients (P = .004), as expected in these patients treated on a phase 1/2 clinical trial before commercial availability of CD19 CAR T-cell products. There was a statistical trend toward older age in tisacel patients (median 64, compared with 62 and 60 in axicel and JCAR014 patients, respectively; P = .08). The median time from leukapheresis to CAR T-cell infusion in axicel, tisacel, and JCAR014 patients was 27, 40, and 17 days, respectively (P < .001) (supplemental Table 2).

Table 1.

Patient and disease characteristics

CharacteristicAxicel, n = 68Tisacel, n = 31JCAR014, n = 30P value*
Age    .077 
 Median (IQR) 62 (50-66) 64 (56-72) 60 (53-64) — 
 Range 25-79 23-81 27-71 — 
Sex, n (%)    .9 
 Male 47 (69) 21 (68) 19 (63) — 
 Female 21 (31) 10 (32) 11 (37) — 
HCT-CI    .5 
 Median (IQR) 1.00 (0.00-3.00) 1.00 (0.00-3.00) 1.00 (0.00-2.75) — 
 Range 0.00-9.00 0.00-6.00 0.00-6.00 — 
Lymphoma histology, n (%)    .004 
 Diffuse large B-cell lymphoma 50 (74) 18 (58) 13 (43) — 
 Transformed 14 (21) 12 (39) 8 (27) — 
 Other large B-cell histologies 4 (5.9) 1 (3.2) 7 (23) — 
 Burkitt lymphoma 0 (0) 0 (0) 2 (6.7) — 
Number of prior therapies    .004 
 Median (IQR) 3.00 (2.00-4.00) 3.00 (2.00-4.00) 4.00 (3.25-5.75) — 
 Range 2.00-9.00 2.00-9.00 2.00-9.00 — 
Relapse pattern, n (%)    .4 
 Relapsed 33 (49) 11 (35) 11 (37) — 
 Secondary refractory 22 (32) 16 (52) 14 (47) — 
 Primary refractory 13 (19) 4 (13) 5 (17) — 
LDH (U/L)    >.9 
 Median (IQR) 231 (168-379) 221 (176-372) 227 (184-307) — 
 Range 105-2812 145-549 110-641 — 
Largest lesion diameter (cm)    .5 
 Median (IQR) 4.60 (2.50-7.30) 4.25 (2.50-6.70) 4.05 (2.58-5.12) — 
 Range 1.10-13.10 1.50-24.80 0.80-10.90 — 
 Missing data 11 — 
Ann Arbor stage, n (%)    .2 
 IV 38 (58) 14 (54) 18 (60) — 
 III 16 (25) 5 (19) 8 (27) — 
 II 11 (17) 4 (15) 4 (13) — 
 I 0 (0) 3 (12) 0 (0) — 
 Missing data — 
Extranodal disease, n (%) 40 (62) 21 (75) 18 (60) .4 
 Missing data — 
ALC (g/L)    .8 
 Median (IQR) 0.72 (0.42-1.10) 0.64 (0.45-0.99) 0.76 (0.53-1.03) — 
 Range 0.11-3.82 0.28-2.69 0.27-2.51 — 
Bridging therapy postleukapheresis, n (%) 40 (59) 22 (71) 6 (20) <.001 
CharacteristicAxicel, n = 68Tisacel, n = 31JCAR014, n = 30P value*
Age    .077 
 Median (IQR) 62 (50-66) 64 (56-72) 60 (53-64) — 
 Range 25-79 23-81 27-71 — 
Sex, n (%)    .9 
 Male 47 (69) 21 (68) 19 (63) — 
 Female 21 (31) 10 (32) 11 (37) — 
HCT-CI    .5 
 Median (IQR) 1.00 (0.00-3.00) 1.00 (0.00-3.00) 1.00 (0.00-2.75) — 
 Range 0.00-9.00 0.00-6.00 0.00-6.00 — 
Lymphoma histology, n (%)    .004 
 Diffuse large B-cell lymphoma 50 (74) 18 (58) 13 (43) — 
 Transformed 14 (21) 12 (39) 8 (27) — 
 Other large B-cell histologies 4 (5.9) 1 (3.2) 7 (23) — 
 Burkitt lymphoma 0 (0) 0 (0) 2 (6.7) — 
Number of prior therapies    .004 
 Median (IQR) 3.00 (2.00-4.00) 3.00 (2.00-4.00) 4.00 (3.25-5.75) — 
 Range 2.00-9.00 2.00-9.00 2.00-9.00 — 
Relapse pattern, n (%)    .4 
 Relapsed 33 (49) 11 (35) 11 (37) — 
 Secondary refractory 22 (32) 16 (52) 14 (47) — 
 Primary refractory 13 (19) 4 (13) 5 (17) — 
LDH (U/L)    >.9 
 Median (IQR) 231 (168-379) 221 (176-372) 227 (184-307) — 
 Range 105-2812 145-549 110-641 — 
Largest lesion diameter (cm)    .5 
 Median (IQR) 4.60 (2.50-7.30) 4.25 (2.50-6.70) 4.05 (2.58-5.12) — 
 Range 1.10-13.10 1.50-24.80 0.80-10.90 — 
 Missing data 11 — 
Ann Arbor stage, n (%)    .2 
 IV 38 (58) 14 (54) 18 (60) — 
 III 16 (25) 5 (19) 8 (27) — 
 II 11 (17) 4 (15) 4 (13) — 
 I 0 (0) 3 (12) 0 (0) — 
 Missing data — 
Extranodal disease, n (%) 40 (62) 21 (75) 18 (60) .4 
 Missing data — 
ALC (g/L)    .8 
 Median (IQR) 0.72 (0.42-1.10) 0.64 (0.45-0.99) 0.76 (0.53-1.03) — 
 Range 0.11-3.82 0.28-2.69 0.27-2.51 — 
Bridging therapy postleukapheresis, n (%) 40 (59) 22 (71) 6 (20) <.001 

JCAR014, defined CD4+/CD8+-composition CD19 CAR T cells.

All variables were assessed before leukapheresis unless specified.

*

Kruskal-Wallis rank-sum test; Fisher's exact test.

Outcomes after CD19 CAR T-cell therapy

Toxicity

Proportions of patients categorized by CRS and ICANS grade and by CAR T-cell product type are shown in Figure 2 and Table 2. We observed higher rates of grade ≥1 CRS in axicel (87%; 95% CI, 76-94) and tisacel (71%; 95% CI, 52-86) patients compared with JCAR014 (47%; 95% CI, 28-66; P < .001) and grade ≥2 CRS with axicel compared with tisacel and JCAR014 (P = .03). We observed significantly higher rates of grade ≥1 (62%; 95% CI, 49-73; P < .001), grade ≥2 (51%; 95% CI, 39-64; P = .002), and grade ≥3 ICANS (29%; 95% CI, 19-42; P = .04) in axicel patients compared with those who received tisacel and JCAR014. A higher proportion of patients requiring tocilizumab was seen in axicel (49%) and tisacel (42%) patients compared with JCAR014 (7%; P < .001), and higher proportion of patients requiring corticosteroids with axicel (60%) compared with tisacel (16%) and JCAR014 (10%; P < .001).

Figure 2.

CRS and ICANS grades across CAR T-cell products. CRS and ICANS were graded using the ASTCT consensus criteria and the CTCAE 4.03, respectively.

Figure 2.

CRS and ICANS grades across CAR T-cell products. CRS and ICANS were graded using the ASTCT consensus criteria and the CTCAE 4.03, respectively.

Close modal
Table 2.

CRS, ICANS, and best response after CD19 CAR T-cell therapy

CharacteristicAxicel, n = 68Ttisacel, n = 31JCAR014, n = 30P value*
CRS, n (%)     
 Grade ≥1 59 (87) 22 (71) 14 (47) <.001 
 Grade ≥2 33 (49) 9 (29) 7 (23) .030 
 Grade ≥3 5 (7.4) 0 (0) 0 (0) .2 
CRS duration, d    .024 
 Median (IQR) 6.0 (4.0-9.0) 5.0 (3.0-6.0) 4.5 (3.0-5.0)  
 Range 2.0-22.0 1.0-14.0 2.0-6.0  
ICANS, n (%)     
 Grade ≥1 42 (62) 7 (23) 6 (20) <.001 
 Grade ≥2 35 (51) 7 (23) 5 (17) <.001 
 Grade ≥3 20 (29) 4 (13) 3 (10) .045 
ICANS duration, d    .2 
 Median (IQR) 6 (3-14) 5 (2-7) 3 (2-4)  
 Range 1-101 1-27 1-5  
Tocilizumab, n (%) 33 (49) 13 (42) 2 (6.7) <.001 
Corticosteroids, n (%) 41 (60) 5 (16) 3 (10) <.001 
Response, n (%)     
 Overall response rate (best response) 49 (75) 18 (58) 17 (57) .10 
 Overall response rate (best response, CT-only criteria) 41 (63) 16 (52) 14 (50) .4 
 Overall response rate (best response, PET-only criteria) 47 (80) 13 (68) 17 (65) .3 
 CR rate (best response) 36 (55) 10 (32) 12 (40) .077 
 CR rate (best response, CT-only criteria) 16 (25) 7 (23) 3 (11) .3 
 CR rate (best response, PET-only criteria) 34 (58) 9 (47) 12 (46) .5 
CharacteristicAxicel, n = 68Ttisacel, n = 31JCAR014, n = 30P value*
CRS, n (%)     
 Grade ≥1 59 (87) 22 (71) 14 (47) <.001 
 Grade ≥2 33 (49) 9 (29) 7 (23) .030 
 Grade ≥3 5 (7.4) 0 (0) 0 (0) .2 
CRS duration, d    .024 
 Median (IQR) 6.0 (4.0-9.0) 5.0 (3.0-6.0) 4.5 (3.0-5.0)  
 Range 2.0-22.0 1.0-14.0 2.0-6.0  
ICANS, n (%)     
 Grade ≥1 42 (62) 7 (23) 6 (20) <.001 
 Grade ≥2 35 (51) 7 (23) 5 (17) <.001 
 Grade ≥3 20 (29) 4 (13) 3 (10) .045 
ICANS duration, d    .2 
 Median (IQR) 6 (3-14) 5 (2-7) 3 (2-4)  
 Range 1-101 1-27 1-5  
Tocilizumab, n (%) 33 (49) 13 (42) 2 (6.7) <.001 
Corticosteroids, n (%) 41 (60) 5 (16) 3 (10) <.001 
Response, n (%)     
 Overall response rate (best response) 49 (75) 18 (58) 17 (57) .10 
 Overall response rate (best response, CT-only criteria) 41 (63) 16 (52) 14 (50) .4 
 Overall response rate (best response, PET-only criteria) 47 (80) 13 (68) 17 (65) .3 
 CR rate (best response) 36 (55) 10 (32) 12 (40) .077 
 CR rate (best response, CT-only criteria) 16 (25) 7 (23) 3 (11) .3 
 CR rate (best response, PET-only criteria) 34 (58) 9 (47) 12 (46) .5 

CRS and ICANS were graded according to the ASTCT consensus criteria and the CTCAE 4.03, respectively. Response after CAR T-cell therapy was assessed using the Lugano 2014 criteria using PET criteria if available or CT criteria in the absence of PET data (n = 21).

*

Fisher’s exact test; Kruskal-Wallis rank-sum test.

Rates of severe infection occurring within 30 days after axicel, tisacel, or JCAR014 were 9%, 3%, and 0%, respectively (P = .20). We observed higher rates of severe neutropenia, thrombocytopenia, and anemia at day 30 in axicel and tisacel patients compared with JCAR014 patients (supplemental Table 3). Early death before disease restaging at day 30 after CAR T-cell infusion was reported in 3 axicel patients (ventricular tachycardia in the context of severe ICANS, ischemic stroke, and neutropenic colitis, n = 1; cytomegalovirus pneumonia complicated with diffuse alveolar hemorrhage and multiorgan failure, n = 1; Escherichia coli septic shock, n = 1).

Best overall and CR rates (Table 2)

The best overall response rates after axicel, tisacel, and JCAR014 were 75% (95% CI, 63-85), 58% (95% CI, 39-75), and 57% (95% CI, 37-75), respectively (P = .10). The complete response rates after axicel, tisacel, and JCAR014 were 55% (95% CI, 43-68), 32% (95% CI, 17-51), and 40% (95% CI, 23-59), respectively (P = .08).

CRS and ICANS severity

Nonadjusted and adjusted odds ratios (ORs) suggested JCAR014 was independently associated with significantly lower CRS severity compared with axicel (adjusted odds ratio [aOR], 0.19; 95% CI, 0.08-0.46; P < .001). The aOR was also compatible with reduced CRS severity with tisacel compared with axicel (aOR, 0.47; 95% CI, 0.21-1.06; P = .07), although we could not exclude a null effect at the 0.05 level. The impact of age on CRS severity was undetermined (aOR per 10-year increase, 0.86; 95% CI, 0.65-1.10; P = .28). Both tisacel and JCAR014 were associated with lower ICANS severity compared with axicel (aORs, 0.17 and 0.17; 95% CI, 0.06-0.48 and 0.06-0.47; P < .001 and P < .001, respectively). The impact of age on ICANS risk was also undetermined (aOR per 10-year increase, 1.30; 95% CI, 0.92-1.70; P = .15).

Efficacy (CR)

Unadjusted and adjusted OR for CR suggested lower antitumor efficacy with tisacel (aOR, 0.23; 95% CI, 0.06-0.78; P = .02) and JCAR014 (aOR, 0.21; 95% CI, 0.06-0.73; P = .01) compared with axicel, as shown in Table 3, using a multivariable model not including interaction terms. The preleukapheresis LDH (aOR, 0.22; 95% CI, 0.10-0.51; P < .001), largest lesion diameter (aOR, 0.29; 95% CI, 0.09-0.87; P = .03), and ALC (aOR, 2.41; 95% CI, 1.22-4.76; P = .01) were also strongly and independently associated with the odds of CR (Figure 3). Since a higher proportion of tisacel patients underwent CT-based restaging alone (39%) compared with axicel (9%) and JCAR014 (10%) patients, we performed sensitivity analyses modeling CR by either CT- or PET-based criteria to assess the impact of imaging type on our estimates. As shown in Table 3, aORs still suggested JCAR014 was associated with lower efficacy (aOR for CT-based CR, 0.25; 95% CI, 0.06-1.06; P = .06; aOR for PET-based CR, 0.30; 95% CI, 0.09-1.00; P = .05). In contrast, the impact of tisacel compared with axicel was undetermined (aOR for CT-based CR, 0.78; 95% CI, 0.24-2.52; P = .68; aOR for PET-based CR, 0.42; 95% CI, 0.11-1.54; P = .19). Additional sensitivity analyses are available in the supplemental Material (supplemental Table 5, addition of bridging therapy to the set of covariates; supplemental Table 6, exclusion of JCAR014 patients; supplemental Table 7, inclusion of prelymphodepletion instead of preleukapheresis ALC).

Table 3.

CAR T-cell product type and outcome prediction

Unadjusted OR95% CIP (> Z )aOR95% CIP (> Z )
CRS grade*       
 Tisacel vs axicel 0.43 0.2-0.95 .04 0.47 .21-1.06 .07 
 JCAR014 vs axicel 0.19 0.08-0.46 <.001 0.19 0.08-0.46 <.001 
ICANS grade*       
 Tisacel vs axicel 0.22 0.08-0.56 .002 0.17 0.06-0.48 <.001 
 JCAR014 vs axicel 0.18 0.07-0.48 <.001 0.17 0.06-0.47 <.001 
CR (best response)       
 Tisacel vs axicel 0.38 0.16-0.94 .04 0.23 0.06-0.78 .02 
 JCAR014 vs axicel 0.54 0.22-1.29 .16 0.21 0.06-0.73 .01 
CR (best response, CT-based criteria only)       
 Tisacel vs axicel 0.89 0.32-2.46 .83 0.78 0.24-2.52 .68 
 JCAR014 vs axicel 0.37 0.1-1.38 .14 0.25 0.06-1.06 .06 
CR (best response, PET-based criteria only)       
 Tisacel vs axicel 0.66 0.23-1.87 .43 0.42 0.11-1.54 .19 
 JCAR014 vs axicel 0.63 0.25-1.59 .33 0.3 0.09-1 .05 
Unadjusted OR95% CIP (> Z )aOR95% CIP (> Z )
CRS grade*       
 Tisacel vs axicel 0.43 0.2-0.95 .04 0.47 .21-1.06 .07 
 JCAR014 vs axicel 0.19 0.08-0.46 <.001 0.19 0.08-0.46 <.001 
ICANS grade*       
 Tisacel vs axicel 0.22 0.08-0.56 .002 0.17 0.06-0.48 <.001 
 JCAR014 vs axicel 0.18 0.07-0.48 <.001 0.17 0.06-0.47 <.001 
CR (best response)       
 Tisacel vs axicel 0.38 0.16-0.94 .04 0.23 0.06-0.78 .02 
 JCAR014 vs axicel 0.54 0.22-1.29 .16 0.21 0.06-0.73 .01 
CR (best response, CT-based criteria only)       
 Tisacel vs axicel 0.89 0.32-2.46 .83 0.78 0.24-2.52 .68 
 JCAR014 vs axicel 0.37 0.1-1.38 .14 0.25 0.06-1.06 .06 
CR (best response, PET-based criteria only)       
 Tisacel vs axicel 0.66 0.23-1.87 .43 0.42 0.11-1.54 .19 
 JCAR014 vs axicel 0.63 0.25-1.59 .33 0.3 0.09-1 .05 
*

From a multivariable proportional odds logistic regression model including the following variables: CAR T-cell product type, preleukapheresis LDH, preleukapheresis largest lesion diameter, age, HCT-CI, preleukapheresis ALC. P values per the Wald test.

From a logistic regression model including the following variables: CAR T-cell product type, preleukapheresis LDH, preleukapheresis largest lesion diameter, age, HCT-CI, preleukapheresis ALC. P values per the Wald test.

Figure 3.

Forest plot of predictors of CR after CD19 CAR T-cell therapy. OR and 95% CI from a multivariable logistic regression model including the following predictors: CAR T-cell product type, preleukapheresis LDH, preleukapheresis largest lesion diameter, age, HCT-CI, and preleukapheresis ALC. For continuous variables, ORs were rescaled to reflect an increase from the first (25th percentile) to the third quartile (75th percentile).

Figure 3.

Forest plot of predictors of CR after CD19 CAR T-cell therapy. OR and 95% CI from a multivariable logistic regression model including the following predictors: CAR T-cell product type, preleukapheresis LDH, preleukapheresis largest lesion diameter, age, HCT-CI, and preleukapheresis ALC. For continuous variables, ORs were rescaled to reflect an increase from the first (25th percentile) to the third quartile (75th percentile).

Close modal

Analyses of CAR T-cell product effect modifiers

We hypothesized that 4-1BB costimulated products (tisacel and JCAR014) might be differentially affected by tumor burden and T-cell function-altering factors compared with the CD28-costimulated product axicel. Hence, we tested for the presence of covariates modifying the effect of the CAR T-cell product type on outcomes (ie, interaction effects). To maximize statistical power, and since our models would not fit with the 3-level CAR T-cell product type variable due to the limited sample size, the tisacel and JCAR014 categories were pooled into a single category to create a 2-level categorical variable (axicel, tisacel/JCAR014). Multivariable models for CRS grade, ICANS grade, and complete response were fitted, including the following interaction terms: CAR T-cell product*preleukapheresis LDH, CAR T-cell product*preleukapheresis largest lesion diameter, CAR T-cell product*age, and CAR T-cell product*HCT-CI.

We could not detect significant associations between the interaction terms listed above and CRS (supplemental Table 6) or ICANS (supplemental Table 7). In contrast, our multivariable modeling suggested an interaction effect between the preleukapheresis ALC (P = .04) and the CAR T-cell product type on the odds of achieving a CR (supplemental Table 8) with a stronger association between ALC and efficacy in axicel patients compared with tisacel/JCAR014 patients. The probabilities of CR as a function of the CAR T-cell product type and preleukapheresis ALC are shown in Figure 4.

Figure 4.

The efficacy of distinct CAR T-cell products is differentially impacted by preleukapheresis ALC. Response probabilities from a multivariable logistic regression model are shown as a function of the preleukapheresis ALC. Shaded areas are 95% CIs of the probability estimates. Estimates computed from a logistic regression model included the following predictors: CAR T-cell product type, preleukapheresis LDH, preleukapheresis largest lesion diameter, age, HCT-CI, preleukapheresis ALC, and the following interaction terms: CAR T-cell product*preleukapheresis ALC, CAR T-cell product*preleukapheresis LDH, CAR T-cell product*preleukapheresis largest lesion diameter, CAR T-cell product*age, and CAR T-cell product*HCT-CI. All probabilities are adjusted to the median values of the remaining covariates included in the model.

Figure 4.

The efficacy of distinct CAR T-cell products is differentially impacted by preleukapheresis ALC. Response probabilities from a multivariable logistic regression model are shown as a function of the preleukapheresis ALC. Shaded areas are 95% CIs of the probability estimates. Estimates computed from a logistic regression model included the following predictors: CAR T-cell product type, preleukapheresis LDH, preleukapheresis largest lesion diameter, age, HCT-CI, preleukapheresis ALC, and the following interaction terms: CAR T-cell product*preleukapheresis ALC, CAR T-cell product*preleukapheresis LDH, CAR T-cell product*preleukapheresis largest lesion diameter, CAR T-cell product*age, and CAR T-cell product*HCT-CI. All probabilities are adjusted to the median values of the remaining covariates included in the model.

Close modal

Single-arm clinical trial data of CD19 CAR T-cell therapy have suggested important variations in the toxicity and efficacy rates across distinct CAR T-cell products. Using data from patients treated with an FDA-approved, commercially available CAR T-cell product (axicel or tisacel) or the investigational product JCAR014, we aimed to estimate the independent impact of the CAR T-cell product type on outcomes. To minimize treatment allocation bias, we used an established causal framework based on a DAG to adjust for the key patient and disease characteristics that might have influenced both the choice of CAR T-cell product and outcomes after CAR T-cell therapy.

We found that axicel was independently associated with higher ICANS severity compared with tisacel and JCAR014 and with higher CRS severity compared with JCAR014. Although retrospective, nonblinded grading of toxicities could have introduced bias, we also observed longer CRS duration and higher corticosteroid use after axicel than tisacel and JCAR014 (Table 2).

As previously described by our group and others, the preleukapheresis LDH4,8 and ALC7 were strong predictors of CR after CD19 CAR T-cell therapy. Furthermore, we detected an interaction effect between the preleukapheresis ALC and the CAR T-cell product type, implying the efficacy of distinct CD19 CAR T-cell products may be differentially affected by factors impacting T-cell function. While higher CR rates were observed and predicted after axicel in preleukapheresis ALChigh patients, lower and comparable efficacy was observed and predicted in ALClow patients across all CAR T-cell products (Figure 4). More research is needed to characterize the mechanisms responsible for this differential impact.

An important limitation of our study is that differences in response assessments may have influenced the observed response rates in our dataset. Specifically, CT-only assessments were performed more frequently in tisacel patients (39%) due to distinct insurance policies between institutions and states for PET imaging coverage, compared with axicel (9%) and JCAR014 (10%) patients. When considering PET criteria or CT criteria alone, the impact of tisacel on the odds of CR compared with axicel became undetermined (Table 3). While this could be related to low power (due to the reduced sample size), this also highlights the need for homogenous response assessments across CAR T-cell products.

In this study, we chose to compare clinical trial patients to real-world patients. It is generally accepted that clinical trial data suffer from the selection of patients with less aggressive disease. In contrast, we found that patients treated with investigational JCAR014 had been more heavily pretreated and that response rates were comparable or inferior to real-world patients.

It is also possible that other confounding factors related to tumor aggressiveness not captured in our dataset may have confounded our estimates. We tried to minimize this bias by adjusting for a measure of both tumor kinetics (preleukapheresis LDH) and bulk (largest lesion diameter before leukapheresis). Recent publications have suggested the total metabolic tumor volume may be a better predictor of outcomes after CD19 CAR T-cell therapy over LDH or tumor bulk.23,24 Unfortunately, the total metabolic tumor volume is not routinely measured at our institutions, and the data were not available to be included in our models. Last, unmeasured confounders, such as physician and patient preferences and insurance coverage, not modeled in our DAG, might have also generated allocation bias. A randomized clinical trial would address this limitation but is not currently forthcoming as it would most likely require the support of nonindustry sponsors such as the National Clinical Trial Network of the National Cancer Institute.

Given significant differences in the manufacturing process between JCAR014 and JCAR017 (JCAR014, antigen presenting cell-based ex vivo stimulation with an infusion of a fresh product containing 2 × 106CAR T cells/kg; JCAR017, cytokine-based ex vivo stimulation with an infusion of a cryopreserved product containing a total dose of 50 to 110 × 106 CAR T cells), our findings should not be extrapolated to compare outcomes between lisocel and other commercially approved CAR T-cell products. A recently published matching-adjusted indirect treatment comparison of lisocel vs axicel suggested comparable efficacy and a more favorable safety profile with lisocel.25 

In conclusion, our analyses show a strong and independent impact of the CAR T-cell product type on toxicity severity and antitumor efficacy. Antitumor efficacy was comparable across all 3 products in preleukapheresis ALClow patients. Higher CR probabilities were observed and predicted after axicel in ALChigh patients compared with tisacel and JCAR014. Further studies are needed to analyze cohorts of patients including other FDA-approved CAR T-cell products (eg, lisocel, brexucabtagene autoleucel) to confirm our findings, pursue comparative studies of health resource utilization, and further optimize patient and CAR T-cell product selection in patients with R/R B-NHL.

The authors thank both the Fred Hutchinson Cancer Research Center Cell Processing Facility and Seattle Cancer Care Alliance Cell Therapy Laboratory, the staff of the Program in Immunology and Seattle Cancer Care Alliance Immunotherapy Clinic, and the Oregon Health and Science University Cell Therapy Clinical, Administrative, and Laboratory Programs.

J.G. thanks the National Institutes of Health/National Cancer Institute Cancer Center Support Grant (P30 CA015704-45).

Contribution: J.G. and R.T.M. conceptualized and designed the study; J.G., N.G., A.V.H., C.J.T., D.G.M., S.W., J.M., and J.A.H. collected and assembled data; J.G., C.J.T., D.G.M., and R.T.M. analyzed and interpreted data; and all authors wrote and approved the final manuscript and are accountable for all aspects of the study.

Conflict-of-interest disclosure: J.G. reports honoraria from Larvol, Eusapharma, JMP, Multerra Bio; research support from Juno Therapeutics, a Bristol Myers Squibb company, and Sobi; and participation in an advisory board for JNJ/Legend Biotech. A.V.H. reports honoraria from Bristol Myers Squibb and Novartis and research support from Juno Therapeutics, a Bristol Myers Squibb company. A. Chen has participated in advisory board meetings for Morphosys, Mesoblast, and Fate Therapeutics. J.A.H. has served as a consultant for Allogene and Gilead, with research support from Gilead. S.N. is employed by Novartis. V.A.C. has received research funding from AstraZeneca. D.G.M. has received research funding paid to his institution and honoraria from Juno Therapeutics, a BMS Company, Celgene, a BMS Company, and Kite Pharma; has participated in ad hoc advisory board meetings and received honoraria from Amgen, BMS, Genentech, Gilead Sciences, Incyte, Janssen, Legend Biotech, Mustang Bio, MorphoSys, Novartis, Pharmacyclics, and Umoja; has rights to royalties from Fred Hutch for patents licensed to Juno/BMS; and is a member of the A2 Biotherapeutics Scientific Advisory Board with stock options and compensation and is a member of the Navan Technologies Scientific Advisory Board with stock options and compensation. C.J.T. receives research funding from Juno Therapeutics/BMS and Nektar Therapeutics; serves on Scientific Advisory Boards for Precision Biosciences, Eureka Therapeutics, Caribou Biosciences, T-CURX, Myeloid Therapeutics, ArsenalBio, and Century Therapeutics; has stock/options in Precision Biosciences, Eureka Therapeutics, Caribou Biosciences, Myeloid Therapeutics, and ArsenalBio; has the right to receive royalties from Fred Hutch as an inventor on patents related to CAR T-cell therapy that are licensed to Juno Therapeutics/BMS; and served on ad hoc advisory boards (last 12 months) for GlaxoSmithKline. M.L.S. is a member of consulting, advisory boards, steering committees, or data safety monitoring committees: Abbvie, Genentech, AstraZeneca, Sound Biologics, Pharmacyclics, Beigene, Bristol Myers Squibb, Morphosys/Incyte, TG Therapeutics, Innate Pharma, Kite Pharma, Adaptive Biotechnologies, Epizyme, Eli Lilly, Adaptimmune, Mustang Bio, Regeneron, Merck, Fate Therapeutics, MEI Pharma, and Atara Biotherapeutics; Research funding: Mustang Bio, Celgene, Bristol Myers Squibb, Pharmacyclics, Gilead, Genentech, AbbVie, TG Therapeutics, Beigene, AstraZeneca, Sunesis, Atara Biotherapeutics, Genmab, and Morphosys/Incyte. R.T.M. is an advisor or consultant for AlloVir, Artiva, CRISPR Therapeutics, CytoDyn, Incyte, and Novartis; reports honoraria from Bristol-Myers Squibb/Celgene, Incyte, Intellia, Kite, Omeros, Orca BioSystems, and PACT Pharma; research support from BMS/Celgene/Juno and Novartis; participation in a data and safety monitoring board for Athersys, Novartis, and Vor Pharmaceutical; and holds a patent with Athersys. The remaining authors declare no competing financial interests.

Correspondence: Jordan Gauthier, Clinical Research Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA 98109; e-mail: jgauthier@fredhutch.org.

Results were presented in part at the 2021 American Society of Clinical Oncology and 2021 European Hematology Association annual meetings.

The online version of this article contains a data supplement.

There is a Blood Commentary on this article in this issue.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

1.
Schuster
SJ
,
Bishop
MR
,
Tam
CS
, et al;
JULIET Investigators
.
Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma
.
N Engl J Med.
2019
;
380
(
1
):
45
-
56
.
2.
Neelapu
SS
,
Locke
FL
,
Bartlett
NL
, et al
.
Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma
.
N Engl J Med.
2017
;
377
(
26
):
2531
-
2544
.
3.
Abramson
JS
,
Palomba
ML
,
Gordon
LI
, et al
.
Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study
.
Lancet.
2020
;
396
(
10254
):
839
-
852
.
4.
Hirayama
AV
,
Gauthier
J
,
Hay
KA
, et al
.
The response to lymphodepletion impacts PFS in patients with aggressive non-Hodgkin lymphoma treated with CD19 CAR T cells
.
Blood.
2019
;
133
(
17
):
1876
-
1887
.
5.
Turtle
CJ
,
Hanafi
LA
,
Berger
C
, et al
.
Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells
.
Sci Transl Med.
2016
;
8
(
355
):
355ra116
.
6.
Wang
M
,
Munoz
J
,
Goy
A
, et al
.
KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma
.
N Engl J Med.
2020
;
382
(
14
):
1331
-
1342
.
7.
Jacobson
CA
,
Hunter
BD
,
Redd
R
, et al
.
Axicabtagene ciloleucel in the non-trial setting: outcomes and correlates of response, resistance, and toxicity
.
J Clin Oncol.
2020
;
38
(
27
):
3095
-
3106
.
8.
Nastoupil
LJ
,
Jain
MD
,
Feng
L
, et al
.
Standard-of-care axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma: results from the US lymphoma CAR T consortium
.
J Clin Oncol.
2020
;
38
(
27
):
3119
-
3128
.
9.
Crump
M
,
Neelapu
SS
,
Farooq
U
, et al
.
Outcomes in refractory diffuse large B-cell lymphoma: results from the international SCHOLAR-1 study
.
Blood.
2017
;
130
(
16
):
1800
-
1808
.
10.
Gauthier
J
,
Turtle
CJ
.
Insights into cytokine release syndrome and neurotoxicity after CD19-specific CAR-T cell therapy
.
Curr Res Transl Med.
2018
;
66
(
2
):
50
-
52
.
11.
Hay
KA
,
Hanafi
LA
,
Li
D
, et al
.
Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy
.
Blood.
2017
;
130
(
21
):
2295
-
2306
.
12.
Gust
J
,
Hay
KA
,
Hanafi
LA
, et al
.
endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells
.
Cancer Discov.
2017
;
7
(
12
):
1404
-
1419
.
13.
Turtle
CJ
,
Hanafi
LA
,
Berger
C
, et al
.
CD19 CAR-T cells of defined CD4+:CD8+  composition in adult B cell ALL patients
.
J Clin Invest.
2016
;
126
(
6
):
2123
-
2138
.
14.
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 Transplant.
2019
;
25
(
4
):
625
-
638
.
15.
Gilead
.
Yescarta and tecartus risk evaluation and mitigation strategies
.
2020
. https://www.yescartatecartusrems.com/. Accessed 1 February 2022.
16.
Novartis
.
Kymriah risk evaluation and mitigation strategies
.
2020
. https://www.kymriah-rems.com/. Accessed 1 February 2022.
17.
Cheson
BD
,
Fisher
RI
,
Barrington
SF
, et al;
Alliance, Australasian Leukaemia and Lymphoma Group
;
Eastern Cooperative Oncology Group
;
European Mantle Cell Lymphoma Consortium
;
Italian Lymphoma Foundation
;
European Organisation for Research
;
Treatment of Cancer/Dutch Hemato-Oncology Group
;
Grupo Español de Médula Ósea
;
German High-Grade Lymphoma Study Group
;
German Hodgkin’s Study Group
;
Japanese Lymphorra Study Group
;
Lymphoma Study Association
;
NCIC Clinical Trials Group
;
Nordic Lymphoma Study Group
;
Southwest Oncology Group
;
United Kingdom National Cancer Research Institute
.
Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification
.
J Clin Oncol.
2014
;
32
(
27
):
3059
-
3068
.
18.
Westreich
D
,
Greenland
S
.
The table 2 fallacy: presenting and interpreting confounder and modifier coefficients
.
Am J Epidemiol.
2013
;
177
(
4
):
292
-
298
.
19.
Shapiro
DD
,
Msaouel
P
.
Causal diagram techniques for urologic oncology research
.
Clin Genitourin Cancer.
2020
;
19
(
3
):
271.e1
-
271.e7
.
20.
VanderWeele
TJ
.
Principles of confounder selection
.
Eur J Epidemiol.
2019
;
34
(
3
):
211
-
219
.
21.
Cinelli
C
,
Forney
A
,
Pearl
J
.
A crash course in good and bad controls
. https://ssrn.com/abstract=3689437. Accessed 1 April 2022. 
22.
Textor
J
,
van der Zander
B
,
Gilthorpe
MS
,
Liskiewicz
M
,
Ellison
GT
.
Robust causal inference using directed acyclic graphs: the R package ‘dagitty’
.
Int J Epidemiol.
2016
;
45
(
6
):
1887
-
1894
.
23.
Vercellino
L
,
Di Blasi
R
,
Kanoun
S
, et al
.
Predictive factors of early progression after CAR T-cell therapy in relapsed/refractory diffuse large B-cell lymphoma
.
Blood Adv.
2020
;
4
(
22
):
5607
-
5615
.
24.
Iacoboni
G
,
Simó
M
,
Villacampa
G
, et al
.
Prognostic impact of total metabolic tumor volume in large B-cell lymphoma patients receiving CAR T-cell therapy
.
Ann Hematol.
2021
;
100
(
9
):
2303
-
2310
.
25.
Maloney
DG
,
Kuruvilla
J
,
Liu
FF
, et al
.
Matching-adjusted indirect treatment comparison of liso-cel versus axi-cel in relapsed or refractory large B cell lymphoma
.
J Hematol Oncol.
2021
;
14
(
1
):
140
.

Supplemental data

Sign in via your Institution