• Race, ethnicity, and BMI did not impact CAR T-cell efficacy or neurotoxicity outcomes.

  • Hispanic patients were more likely to experience severe cytokine release syndrome following CAR T-cell therapy.

Cancer outcomes with chemotherapy are inferior in patients of minority racial/ethnic groups and those with obesity. Chimeric antigen receptor (CAR) T-cell therapy has transformed outcomes for relapsed/refractory hematologic malignancies, but whether its benefits extend commensurately to racial/ethnic minorities and patients with obesity is poorly understood. With a primary focus on patients with B-cell acute lymphoblastic leukemia (B-ALL), we retrospectively evaluated the impact of demographics and obesity on CAR T-cell therapy outcomes in adult and pediatric patients with hematologic malignancies treated with CAR T-cell therapy across 5 phase 1 clinical trials at the National Cancer Institute from 2012 to 2021. Among 139 B-ALL CAR T-cell infusions, 28.8% of patients were Hispanic, 3.6% were Black, and 29.5% were overweight/obese. No significant associations were found between race, ethnicity, or body mass index (BMI) and complete remission rates, neurotoxicity, or overall survival. Hispanic patients were more likely to experience severe cytokine release syndrome compared with White non-Hispanic patients even after adjusting for leukemia disease burden and age (odds ratio, 4.5; P = .001). A descriptive analysis of patients with multiple myeloma (n = 24) and non-Hodgkin lymphoma (n = 23) displayed a similar pattern to the B-ALL cohort. Our findings suggest CAR T-cell therapy may provide substantial benefit across a range of demographics characteristics, including for those populations who are at higher risk for chemotherapy resistance and relapse. However, toxicity profiles may vary. Therefore, efforts to improve access to CAR therapy for underrepresented populations and elucidate mechanisms of differential toxicity among demographic groups should be prioritized.

Over the past decade, chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment landscape for multiply relapsed or chemotherapy-refractory hematologic malignancies.1 As a testament to this success, there are currently 6 unique Food and Drug Administration–approved CAR T-cell constructs available for use in relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL),2 B-cell non-Hodgkin lymphoma (NHL),3,4 and multiple myeloma (MM).5,6 Despite the success of CAR T-cell therapy, whether its considerable benefits extend commensurately to high-risk demographic subgroups such as racial/ethnic minorities and patients with obesity is poorly understood. The poor representation of racial and ethnic minorities in early-phase and pivotal CAR T-cell trials has limited this analysis.7,8 

Cancer outcomes, particularly with conventional therapies, are worse in certain racial/ethnic minority populations and those with obesity. Hispanic patients with B-ALL experience higher mortality,9 greater toxicity, and more chemotherapy complications than non-Hispanic patients.10,11 Black race12 and Hispanic ethnicity are independent adverse prognostic factors for B-ALL survival.13 High-risk cytogenetics (eg, IgH-CRLF2 and IKZF1) conferring chemotherapeutic resistance are disproportionally found in Hispanic patients with B-ALL.14 In adults with NHL, analysis of the Surveillance, Epidemiologic, and End Results database from 1997 to 2015 demonstrated that Black patients experience worse survival outcomes compared with White patients despite decreasing incidence of disease overall.15 Among patients diagnosed with MM, Black patients have worse survival outcomes compared with White patients even after controlling for receipt of triplet induction therapy and autologous stem cell transplant.16 

Another risk factor impacting cancer outcomes is obesity. Childhood obesity is associated with increased risk of developing B-ALL, relapsing, and having chemotherapy-resistant disease.17,18 Patients with B-ALL and obesity also have a higher prevalence of high-risk cytogenetics such as CRLF2 rearrangements,19 which is further amplified among Hispanic patients.17 Moreover, obesity independently predicts lower event-free survival rates in pediatric B-ALL.20 In adults, obesity has been shown to increase the risk of developing MM21 and independently increase mortality risk in patients with NHL.22 

Whether similar disparities in outcome for racial/ethnic minorities and patients with obesity extend to immunotherapy is less well understood. Some studies support a more favorable response in adult patients with obesity receiving immunotherapy,23,24 particularly with checkpoint inhibitor therapy,25 but data on adoptive cell therapy are limited.20,26 A recent study demonstrated an association between visceral adiposity and higher body mass index (BMI) with severe cytokine release syndrome (CRS) following CD19 CAR T-cell therapy.27 Given the ability of CAR T cells to overcome chemotherapy resistance, which minority and obese populations may be particularly at risk for, we hypothesized that clinical responses may be similar across race and ethnicity and sought to explore the intersection with obesity. Accordingly, we evaluated the impact of race, ethnicity, and obesity on CAR T-cell therapy efficacy and toxicity outcomes across 5 early-phase clinical trials encompassing 4 distinct CAR constructs and 3 hematologic malignancies in pediatric and adult patients.

Study design

This retrospective study included 5 phase 1 CAR T-cell trials at the National Cancer Institute (NCI). All patients had B-cell malignancies, including B-ALL, NHL, and MM, treated with CAR T-cell therapy between 2012 and 2021. CAR constructs included CD19, CD22, CD19/22 bispecific, and B-cell maturation antigen (BCMA). The CD19 and BCMA CAR constructs used a CD28 costimulatory domain, whereas the CD22 and CD19/22 bispecific CAR constructs had a 4-1BB domain. All individual protocols were approved by the NCI Institutional Review Board, including the retrospective study for this analysis, which is registered at www.clinicaltrials.gov as #NCT03827343. The study was conducted according to the Declaration of Helsinki.

Patient demographics, including sex, race, ethnicity, and pretreatment characteristics, such as BMI and disease burden, were verified by study investigators and collected for analysis from the electronic medical record. Home ZIP codes were used to assess geographic referral patterns as our center is a quaternary and federally funded institution and receives patient referrals from across the world. Race and ethnicity were classified into 3 categories: White non-Hispanic, Hispanic, and all other non-Hispanic. Obesity, overweight, normal weight, and underweight were defined as BMI ≥ 30, 25 to <30, 18.5 to <25, and <18.5 kg/m2, respectively, in adults aged ≥20 and as BMI ≥ 95th, 85th to <95th, 5th to <85th, and <5th percentiles, respectively, in pediatric patients aged 2 to <20 per Centers for Disease Control and Prevention guidelines. BMI was calculated prior to lymphodepleting chemotherapy for all patients. Baseline disease burden in the bone marrow for patients with B-ALL was defined as M1 (<5% blasts), M2 (5% to 25%), and M3 (>25%), with all patients required to have detectable disease at infusion.

Outcomes of interest

The primary objective was interrogating the association of race/ethnicity and BMI with CAR T-cell therapy efficacy and toxicity in patients with B-ALL. Efficacy was measured by the complete remission (CR) rate. In B-ALL, CR was defined as negative measurable residual disease (<0.01% blasts) in the bone marrow 28 days after CAR T-cell infusion. Toxicity outcomes were evaluated by maximum severity of CRS, as graded by the American Society for Transplantation and Cellular Therapy (ASTCT) consensus guidelines,28 and neurotoxicity, recorded by its presence or absence given our inability to reconcile grading criteria across the treatment period with more recent ASTCT guidelines. Secondary objectives included evaluation of representation across demographic subgroups, examining geographic referral patterns, and identifying the association of race/ethnicity and BMI with overall survival in patients with B-ALL treated with CAR T-cell therapy. Because of the small number of patients with NHL or MM, CAR T-cell efficacy and toxicity were descriptively analyzed for these groups. In NHL and MM, CR was defined per the Cheson criteria29 and International Myeloma Working Group Uniform Response Criteria,30 respectively.

Statistical analysis

Patient, disease, and treatment characteristics were summarized with descriptive statistics. Univariate logistic regression was employed to evaluate the association of demographic factors with efficacy and toxicity outcomes. Age was treated as a continuous variable and also dichotomized as adult (age ≥18) vs pediatric (age <18). All outcomes were dichotomized as present vs absent (CR, neurotoxicity) and grade 3 to 5 vs grade 0 to 2 (CRS). For select outcomes, multivariable logistic regression models were generated incorporating parameters using backward selection (retention criteria, P < .05). The number of variables in the final model was limited to prevent overfitting per number of outcome events. Survival was calculated using the Kaplan-Meier method from date of last CAR T-cell infusion to date of death or last follow-up with censor date of August 9, 2021. All statistics were performed in Prism (GraphPad) and SAS Version 9.4.

Patient characteristics and demographics (all subjects)

Our analysis included a total of 186 unique infusions of CAR T-cell therapy over the study period. This included 139 (74.7%) infusions for B-ALL (used for primary analysis), 23 (12.4%) infusions for NHL, and 24 (12.9%) infusions for MM. Reinfusion strategies were excluded from this analysis. Eleven patients received 2 different products at 2 different timepoints. These infusions were considered independent, as interim treatment, disease characteristics, and apheresis materials differed between the 2 infusions (supplemental Table 1). Geographically, 120 (64.5%) patients were referred from within the United States, encompassing 32 states and the District of Columbia (Figure 1A); the remaining 55 (35.5%) patients originated from outside the United States (Figure 1B).

Figure 1.

Map of geographic referral pattern. (A) Geographic origin of 120 domestic patients using ZIP codes. (B) Geographic origin of 175 patients. Eleven patients who were treated with 2 CAR constructs at separate times (for a total of 186 treatments) were counted once for geographic tallying. Maps generated at mapchart.net.

Figure 1.

Map of geographic referral pattern. (A) Geographic origin of 120 domestic patients using ZIP codes. (B) Geographic origin of 175 patients. Eleven patients who were treated with 2 CAR constructs at separate times (for a total of 186 treatments) were counted once for geographic tallying. Maps generated at mapchart.net.

Close modal

Cohort with acute lymphoblastic leukemia (n = 139 infusions)

Patient characteristics and demographics

Among 139 patients infused for B-ALL, median age was 15.1 years (interquartile range [IQR], 9.6-21.2), and 98 (70.5%) patients were male. This cohort included a total of 40 (28.8%) Hispanic patients and 5 (3.6%) Black patients. With respect to BMI, 41 (29.5%) patients were overweight or obese (Table 1). Age and BMI showed a moderate positive correlation (supplemental Figure 1). Median age and sex distribution were similar in non-Hispanic patients compared with Hispanic patients. Although a higher percentage of Hispanic patients were obese (22.5%) compared with non-Hispanic patients (12.1%), the percentage of Hispanic patients classified as overweight or obese (32.5%) was similar to non-Hispanic patients (28.3%) (Table 2).

Table 1.

Patient characteristics

B-ALL (n = 139)MM (n = 24)NHL (n = 23)
Median age, y (IQR) 15.10 (9.55, 21.20) 54.50 (52.75, 59.25) 54.00 (45.50, 64.00) 
Male (%) 98 (70.5) 12 (50.0) 13 (56.5) 
Race/ethnicity (%) 
White (non-Hispanic) 77 (55.4) 17 (70.8) 19 (82.6) 
Hispanic 40 (28.8) 3 (12.5) 2 (8.7) 
Asian 14 (10.1) 0 (0.0) 1 (4.3) 
Black 5 (3.6) 4 (16.7) 1 (4.3) 
Hawaiian/Pacific Islander 1 (0.7) 
Multiracial 1 (0.7) 
Unknown 1 (0.7) 
BMI (%) 
Underweight 7 (5.0) 
Normal weight 91 (65.5) 7 (29.2) 10 (43.5) 
Overweight 20 (14.4) 6 (25.0) 7 (30.4) 
Obese 21 (15.1) 11 (45.8) 6 (26.1) 
B-ALL (n = 139)MM (n = 24)NHL (n = 23)
Median age, y (IQR) 15.10 (9.55, 21.20) 54.50 (52.75, 59.25) 54.00 (45.50, 64.00) 
Male (%) 98 (70.5) 12 (50.0) 13 (56.5) 
Race/ethnicity (%) 
White (non-Hispanic) 77 (55.4) 17 (70.8) 19 (82.6) 
Hispanic 40 (28.8) 3 (12.5) 2 (8.7) 
Asian 14 (10.1) 0 (0.0) 1 (4.3) 
Black 5 (3.6) 4 (16.7) 1 (4.3) 
Hawaiian/Pacific Islander 1 (0.7) 
Multiracial 1 (0.7) 
Unknown 1 (0.7) 
BMI (%) 
Underweight 7 (5.0) 
Normal weight 91 (65.5) 7 (29.2) 10 (43.5) 
Overweight 20 (14.4) 6 (25.0) 7 (30.4) 
Obese 21 (15.1) 11 (45.8) 6 (26.1) 

Patient characteristics for all patients.

Table 2.

B-ALL patient characteristics by race and ethnicity (n = 139)

White non-Hispanic (n = 77)Hispanic (n = 40)Other non-Hispanic (n = 22)
Median age, y (IQR) 15.30 (10.70, 20.30) 15.60 (9.17, 21.38) 12.90 (7.03, 20.35) 
Male (%) 57 (74.0) 29 (72.5) 12 (54.5) 
BMI (%)  
Underweight 5 (6.5) 2 (9.1) 
Normal weight 51 (66.2) 27 (67.5) 13 (59.1) 
Overweight 12 (15.6) 4 (10.0) 4 (18.2) 
Obese 9 (11.7) 9 (22.5) 3 (13.6) 
Disease burden (%)  
M1 28 (36.4) 13 (32.5) 6 (27.3) 
M2 8 (10.4) 5 (12.5) 3 (13.6) 
M3 41 (53.2) 22 (55.0) 13 (59.1) 
Prior allo-HSCT (%) 52 (67.5) 14 (35.0) 11 (50.0) 
Prior CART (%) 35 (45.5) 14 (35.0) 6 (27.3) 
White non-Hispanic (n = 77)Hispanic (n = 40)Other non-Hispanic (n = 22)
Median age, y (IQR) 15.30 (10.70, 20.30) 15.60 (9.17, 21.38) 12.90 (7.03, 20.35) 
Male (%) 57 (74.0) 29 (72.5) 12 (54.5) 
BMI (%)  
Underweight 5 (6.5) 2 (9.1) 
Normal weight 51 (66.2) 27 (67.5) 13 (59.1) 
Overweight 12 (15.6) 4 (10.0) 4 (18.2) 
Obese 9 (11.7) 9 (22.5) 3 (13.6) 
Disease burden (%)  
M1 28 (36.4) 13 (32.5) 6 (27.3) 
M2 8 (10.4) 5 (12.5) 3 (13.6) 
M3 41 (53.2) 22 (55.0) 13 (59.1) 
Prior allo-HSCT (%) 52 (67.5) 14 (35.0) 11 (50.0) 
Prior CART (%) 35 (45.5) 14 (35.0) 6 (27.3) 

Treatment characteristics and disease burden

At the time of CAR T-cell treatment at NCI, patients in our study had received a median of 5 (IQR, 3-6) prior therapy regimens, not including allogenic hematopoietic stem cell transplantation (allo-HSCT). In addition, 55 (39.6%) patients had received prior CAR T-cell therapy and 77 (55.3%) had received at least 1 allo-HSCT. However, a significantly lower proportion of Hispanic (35%; Table 2) and overweight/obese patients (34.1%; Table 3) received allo-HSCT compared with non-Hispanic (63.6%; P = .004) and nonoverweight/obese patients (64.3%; P = .002). Baseline disease burden in the bone marrow was M1 for 47 (33.8%), M2 for 16 (11.5%), and M3 for 76 (54.7%) patients and was similar across race, ethnicity (Table 2), and BMI (Table 3). Across trials, 50 (36.0%) patients received CD19 CAR, 71 (51.1%) received CD22 CAR, and 18 (12.9%) received CD19/22 bispecific CAR (Table 4).

Table 3.

B-ALL patient characteristics by BMI (n = 139)

Not overweight/obese (n = 98)Overweight/obese (n = 41)
Median age, y (IQR) 15.25 (9.70, 20.38) 14.70 (8.40, 22.80) 
Male (%) 67 (68.4) 31 (75.6) 
Race/ethnicity  
White non-Hispanic (%) 56 (57.1) 21 (51.2) 
Hispanic (%) 27 (27.6) 13 (31.7) 
Other non-Hispanic (%) 15 (15.3) 7 (17.1) 
Disease burden (%)  
M1 34 (34.7) 13 (31.7) 
M2 12 (12.2) 4 (9.8) 
M3 52 (53.1) 24 (58.5) 
Prior allo-HSCT (%) 63 (64.3) 14 (34.1) 
Prior CART (%) 42 (42.9) 13 (31.7) 
Not overweight/obese (n = 98)Overweight/obese (n = 41)
Median age, y (IQR) 15.25 (9.70, 20.38) 14.70 (8.40, 22.80) 
Male (%) 67 (68.4) 31 (75.6) 
Race/ethnicity  
White non-Hispanic (%) 56 (57.1) 21 (51.2) 
Hispanic (%) 27 (27.6) 13 (31.7) 
Other non-Hispanic (%) 15 (15.3) 7 (17.1) 
Disease burden (%)  
M1 34 (34.7) 13 (31.7) 
M2 12 (12.2) 4 (9.8) 
M3 52 (53.1) 24 (58.5) 
Prior allo-HSCT (%) 63 (64.3) 14 (34.1) 
Prior CART (%) 42 (42.9) 13 (31.7) 
Table 4.

CAR treatment, toxicity, and response of patients with B-ALL

n (%)Overall (n = 139)Overweight/obese (n = 41)Hispanic (n = 40)Other non-Hispanic (n = 22)
CAR construct 
CD19 50 (36.0) 15 (36.6) 18 (45.0) 7 (31.8) 
CD22 71 (51.1) 19 (46.3) 15 (37.5) 12 (54.5) 
CD19/22 18 (12.9) 7 (17.1) 7 (17.5) 3 (13.6) 
CRS ASTCT grade 
None 33 (23.7) 10 (24.4) 12 (30.0) 6 (27.3) 
47 (33.8) 17 (41.5) 9 (22.5) 6 (27.3) 
34 (24.5) 7 (17.1) 7 (17.5) 6 (27.3) 
20 (14.4) 6 (14.6) 10 (25.0) 3 (13.6) 
5 (3.6)  1 (2.4) 2 (5.0) 1 (4.5) 
Developed neurotoxicity 29 (20.9) 8 (19.5) 9 (22.5) 2 (9.1) 
CR 
Yes 94 (67.6) 26 (63.4) 25 (62.5) 14 (63.6) 
No 44 (31.7) 15 (36.6) 15 (37.5) 7 (31.8) 
Not evaluable 1 (0.7)  1 (4.5) 
n (%)Overall (n = 139)Overweight/obese (n = 41)Hispanic (n = 40)Other non-Hispanic (n = 22)
CAR construct 
CD19 50 (36.0) 15 (36.6) 18 (45.0) 7 (31.8) 
CD22 71 (51.1) 19 (46.3) 15 (37.5) 12 (54.5) 
CD19/22 18 (12.9) 7 (17.1) 7 (17.5) 3 (13.6) 
CRS ASTCT grade 
None 33 (23.7) 10 (24.4) 12 (30.0) 6 (27.3) 
47 (33.8) 17 (41.5) 9 (22.5) 6 (27.3) 
34 (24.5) 7 (17.1) 7 (17.5) 6 (27.3) 
20 (14.4) 6 (14.6) 10 (25.0) 3 (13.6) 
5 (3.6)  1 (2.4) 2 (5.0) 1 (4.5) 
Developed neurotoxicity 29 (20.9) 8 (19.5) 9 (22.5) 2 (9.1) 
CR 
Yes 94 (67.6) 26 (63.4) 25 (62.5) 14 (63.6) 
No 44 (31.7) 15 (36.6) 15 (37.5) 7 (31.8) 
Not evaluable 1 (0.7)  1 (4.5) 

One patient died of toxicity with grade 4 CRS and grade 5 capillary leak syndrome.

Toxicity

Overall, 25 (18%) patients experienced grade ≥3 CRS (Table 4). Hispanic patients had >3 times greater odds of experiencing grade ≥3 CRS compared with White non-Hispanic patients (odds ratio [OR], 3.24; 95% confidence interval [CI], 1.23-8.54; P = .001). However, other non-Hispanic patients had similar odds of grade ≥3 CRS compared with White non-Hispanic patients (OR, 1.68; 95% CI, 0.46-6.08; P = .43). Similarly, the incidence of grade ≥3 CRS did not differ between females vs males (OR, 0.71; 95% CI, 0.261.94; P = .50) or between patients who were overweight/obese vs those who were not (OR, 0.92; 95% CI, 0.35-2.39; P = .86; Figure 2A). Severe CRS toxicity was more common in obese compared with overweight patients (supplemental Table 2). A sensitivity analysis of obese vs nonobese patients showed similar results (Table 5). However, continuous BMI measured in kg/m2 was positively associated with grade ≥3 CRS (OR, 1.1; 95% CI, 1.02-1.18; P = .01; supplemental Table 3).

Figure 2.

Forest plots of toxicity and efficacy by race, ethnicity, sex, and BMI among patients with B-ALL. (A) ORs of severe CRS. (B) ORs of neurotoxicity. (C) ORs of CR. ORs were calculated from simple (univariate) logistic regression models with intercept term. Interval bands represent 95% CIs for the point estimates. Arrowheads indicate CI extending beyond axis range. Overweight/obese (O/O) defined as BMI ≥ 25 in patients aged ≥20 years and BMI ≥ 85th percentile in patients aged 2 to 20.

Figure 2.

Forest plots of toxicity and efficacy by race, ethnicity, sex, and BMI among patients with B-ALL. (A) ORs of severe CRS. (B) ORs of neurotoxicity. (C) ORs of CR. ORs were calculated from simple (univariate) logistic regression models with intercept term. Interval bands represent 95% CIs for the point estimates. Arrowheads indicate CI extending beyond axis range. Overweight/obese (O/O) defined as BMI ≥ 25 in patients aged ≥20 years and BMI ≥ 85th percentile in patients aged 2 to 20.

Close modal
Table 5.

Odds of CRS (grade 3 to 5 vs 0 to 2) in B-ALL (n = 139)

Variable (risk group vs ref)UnivariateMultivariable
OR (95% CI)POR (95% CI)P
Age (continuous, y) 1.13 (1.06-1.21) <.001 1.15 (1.07-1.23) <.001 
≥18 y (adult) vs <18 y (pediatric) 4.94 (1.90-12.8) .001 — — 
Sex (male vs female) 1.40 (0.52-3.82) .51 — — 
Race/ethnicity (Hispanic vs White NH) 3.24 (1.23-8.54) .02 4.51 (1.46-13.9) .001 
Other non-Hispanic vs White NH 1.68 (0.46-6.08) .43 2.0 (0.48-8.41) .34 
Prior allo-HSCT (yes vs no) 0.38 (0.16-0.93) .04 — — 
Prior CART (yes vs no) 0.42 (0.16-1.13) .08 — — 
Disease burden (M2/3 vs M1) 4.61 (1.30-16.3) .02 5.48 (1.40-21.5) .01 
BMI (O/O vs non-O/O) 0.92 (0.35-2.40) .86 — — 
Obese vs nonobese 2.1 (0.72-6.1) .18 — — 
CAR (CD22 vs CD19) 0.93 (0.36-2.4) .88 — — 
CD19/22 vs CD19 1.30 (0.35-4.90) .7 — — 
Variable (risk group vs ref)UnivariateMultivariable
OR (95% CI)POR (95% CI)P
Age (continuous, y) 1.13 (1.06-1.21) <.001 1.15 (1.07-1.23) <.001 
≥18 y (adult) vs <18 y (pediatric) 4.94 (1.90-12.8) .001 — — 
Sex (male vs female) 1.40 (0.52-3.82) .51 — — 
Race/ethnicity (Hispanic vs White NH) 3.24 (1.23-8.54) .02 4.51 (1.46-13.9) .001 
Other non-Hispanic vs White NH 1.68 (0.46-6.08) .43 2.0 (0.48-8.41) .34 
Prior allo-HSCT (yes vs no) 0.38 (0.16-0.93) .04 — — 
Prior CART (yes vs no) 0.42 (0.16-1.13) .08 — — 
Disease burden (M2/3 vs M1) 4.61 (1.30-16.3) .02 5.48 (1.40-21.5) .01 
BMI (O/O vs non-O/O) 0.92 (0.35-2.40) .86 — — 
Obese vs nonobese 2.1 (0.72-6.1) .18 — — 
CAR (CD22 vs CD19) 0.93 (0.36-2.4) .88 — — 
CD19/22 vs CD19 1.30 (0.35-4.90) .7 — — 

Association of patient, treatment, and disease characteristics with CRS severity. Univariate and multivariable ORs were calculated using logistic regression models. The significance of the ‘bold’ font is to signify those which have a significant p-value. The “dash” indicates that the particular variable that it was associated with was not carried forward to the multivariable analysis.

To further investigate the association between Hispanic ethnicity and severe CRS, we evaluated the relationship between additional factors and CRS severity. Although baseline disease burden in the bone marrow did not differ significantly by ethnicity (Table 2), high (M2/M3) disease burden was found to be associated with a higher incidence of grade ≥3 CRS compared with low (M1) disease burden (OR, 4.61; 95% CI, 1.30-16.3; P = .02), in line with previous studies examining clinical correlates of CAR-related toxicity.31 Interestingly, we also found that adult patients with B-ALL had nearly 5 times greater odds experiencing grade ≥3 CRS compared with pediatric patients with B-ALL (OR, 4.94; 95% CI, 1.90-12.9; P = .001). Multivariable regression revealed that in a model accounting for age, race/ethnicity, and disease burden, all 3 variables were independently associated with higher odds of severe CRS (C-statistic = 0.82; Table 5). In a second multivariable model, continuous BMI measured in kg/m2 was not significantly associated with CRS severity after adjusting for age, ethnicity, and disease burden (supplemental Table 3). Strikingly, Hispanic patients who were overweight or obese had the greatest odds of severe CRS (Figure 3).

Figure 3.

CRS severity stratified by ethnicity and BMI. Statistical comparison performed using χ2 test. NH, non-Hispanic; OW, overweight.

Figure 3.

CRS severity stratified by ethnicity and BMI. Statistical comparison performed using χ2 test. NH, non-Hispanic; OW, overweight.

Close modal

To verify that differences in CRS severity were not due to differences in CRS incidence overall, we performed a sensitivity analysis with CRS dichotomized as grade 1 to 5 vs grade 0. No differences in CRS incidence were found between groups. Furthermore, neither CAR T-cell construct (P > .7) nor CAR T-cell dose (P = .96) was associated with CRS severity in our study. Finally, as the management of CRS has shifted over the years to earlier and more frequent administration of tocilizumab,32 we evaluated whether incidence of severe CRS in our study was related to treatment year; no relationship was observed for all patients with B-ALL (supplemental Figure 2). Among Hispanic patients with B-ALL, the incidence of severe CRS trended upward over time (supplemental Figure 3).

Neurotoxicity occurred in 29 (20.9%) patients (Table 4). Hispanic patients did not differ with respect to incidence of neurotoxicity compared with White non-Hispanic patients (OR, 0.95; 95% CI, 0.38-2.34; P = .92). Similarly, the odds of neurotoxicity in other non-Hispanic patients were statistically comparable to White non-Hispanic patients (OR, 0.33; 95% CI, 0.07-1.54; P = .16). In contrast, males had >3 times greater odds than females for neurotoxicity (OR, 3.17; 95% CI, 1.03-9.78; P = .05). Neurotoxicity incidence was similar between patients who were overweight/obese and those who were not (OR, 0.89; 95% CI, 0.36-2.21; P = .80; Figure 2B). Other patient characteristics, including baseline disease burden, age, and CAR T-cell construct, were not associated with incidence of neurotoxicity (supplemental Table 4).

Efficacy

Overall, 94 (67.6%) patients achieved CR (Table 4). CR rates were comparable between Hispanic vs White non-Hispanic patients (OR, 0.67; 95% CI, 0.30-1.50; P = .33); other non-Hispanic vs White non-Hispanic patients (OR, 0.81; 95% CI, 0.29-2.25; P = .67); males vs females (OR, 0.60; 95% CI, 0.26-1.37; P = .22); and patients who were overweight/obese vs those who were not (OR, 0.74; 95% CI, 0.34-1.60; P = .44; Figure 2C). Further analysis revealed that only M2/M3 vs M1 disease burden was well associated with lower CR rates (OR, 0.38; 95% CI, 0.16-0.88; P = .02; supplemental Table 5).

Because of the limited representation of Black and African American patients in our trials, which impaired our ability to perform inferential statistical comparisons for this specific group, we descriptively evaluated outcomes of the 5 Black patients with B-ALL in our study. Four (80%) patients achieved CR, including 1 patient with M3 disease burden, and none experienced neurotoxicity or grade ≥3 CRS. Among 14 Asian patients, 7 (50%) achieved CR, 2 (14.3%) experienced grade ≥ 3 CRS, and 1 (7.1%) experienced neurotoxicity.

Overall survival (n = 128)

Median OS was 12.2 months (Figure 4A). However, no statistically significant differences in survival were observed between male vs female patients (11.8 vs 14.9 months; P = .66; Figure 4B); Hispanic vs White non-Hispanic patients (14.6 vs 12.8 months; Figure 4C); other non-Hispanic vs White non-Hispanic patients (12.8 vs 7.9 months; Figure 4C); or overweight/obese patients vs nonoverweight/obese patients (14.9 vs 11.9 months; P = .28; Figure 4D). By the end of the study period, 3 (60%) Black patients (supplemental Figure 4) and 10 (71.4%) Asian patients had died.

Figure 4.

Kaplan-Meier survival curves of patients with B-ALL by race, ethnicity, sex, and BMI. (A) Survival of entire B-ALL cohort. (B) Survival comparison by sex. (C) Survival comparison by race/ethnicity. (D) Survival comparison by BMI. P values calculated using log rank method. t = 0 corresponds to time of last CAR T-cell infusion. Overweight/obese (O/O) defined as BMI ≥ 25 in patients age ≥20 years and BMI ≥ 85th percentile in patients aged 2 to 20.

Figure 4.

Kaplan-Meier survival curves of patients with B-ALL by race, ethnicity, sex, and BMI. (A) Survival of entire B-ALL cohort. (B) Survival comparison by sex. (C) Survival comparison by race/ethnicity. (D) Survival comparison by BMI. P values calculated using log rank method. t = 0 corresponds to time of last CAR T-cell infusion. Overweight/obese (O/O) defined as BMI ≥ 25 in patients age ≥20 years and BMI ≥ 85th percentile in patients aged 2 to 20.

Close modal

Cohort with MM (n = 24)

All MM patients received BCMA CAR. Six (25%) experienced severe CRS, including 1 (33%) Hispanic patient and 4 (23.5%) overweight/obese patients. Neurotoxicity occurred in 3 (12.5%) patients, none of whom were Hispanic and all of whom were overweight/obese. Only 2 (8.3%) patients with MM achieved CR, one of whom was overweight/obese and one of whom was Black (nonoverweight/obese) (supplemental Table 6). Among the 4 Black patients, none experienced severe CRS or any grade of neurotoxicity.

Cohort with NHL (n = 23)

All but 1 NHL patient received CD19 CAR therapy. Severe CRS toxicity occurred in 6 (26.1%) of the NHL patients, including 1 (50%) Hispanic patient and 5 (38.5%) overweight/obese patients. Three (13%) patients, all of whom were overweight/obese and one of whom was Hispanic, experienced neurotoxicity. Ten (43.5%) patients achieved CR, including 1 (50%) Hispanic and 7 (53.8%) overweight/obese patients (supplemental Table 6). One Black patient with Burkitt lymphoma treated on the CD19/22 CAR T-cell trial33 did not achieve CR nor experienced neurotoxicity or CRS and died of progressive disease.

CAR T-cell therapy has transformed outcomes for relapsed/refractory hematologic malignancies, yet whether its benefits apply equally to diverse patient populations and patients with common comorbidities, such as obesity, is poorly understood. Given the tremendous potential of CAR T-cell therapy to overcome chemotherapy-resistant and refractory disease, understanding whether its benefits extend equally to racial and ethnic minorities and patients with obesity is of critical importance to assessing the generalizability of existing outcome data for this novel cancer treatment modality and potentially improving outcomes for the highest-risk populations.

One of the overarching challenges in addressing this question has been the underrepresentation of minority populations in both early-phase clinical trials and pivotal registration studies.34 In 2 landmark phase 3 trials in the United States, representing the very first randomized studies of CAR T-cell therapy to date, ∼80% of patients were White and <10% were Hispanic or Black, a stark contrast from the current demographic landscape.3,35 Despite our center being a federally funded institution that enrolls children and young adults on clinical trials independent of insurance coverage with wide national and international recruitment, our study also shows underrepresentation of Black patients, limiting our ability to discern outcomes by race. This is consistent with generally lower enrollment of Black Americans in cancer clinical trials,36 particularly on phase 1 trials where efficacy is not a primary objective, which poses a critical additional barrier.37 Understanding and overcoming barriers to enrollment of minority populations remains an active effort.

Recent retrospective studies have attempted to address this gap in knowledge. A study of 185 patients with B-ALL treated with commercial CD19 CAR therapy (tisagenlecleucel) demonstrated that Black children and young adults (n = 11, 5.9%) were less likely to receive CAR infusion and had worse survival outcomes compared with non-Black patients.26 On the other hand, a study of 78 adult patients with NHL showed no differences in toxicity, efficacy, or survival between obese and nonobese patients receiving CD19 CAR T-cell therapy.38 Our study, which examined the impact of race, ethnicity, and obesity on CAR T-cell toxicity and efficacy outcomes in both adult and pediatric patients with diverse geographic backgrounds across multiple hematologic malignancies and CAR T-cell constructs, serves to add to these limited data.

We show that CR rates with CAR T-cell therapy in this heavily pretreated population were high in B-ALL but limited in NHL and MM, accounting for the phase I and dose-escalation nature of our trials.39-43 Importantly, no differences in CR rates were observed by sex, ethnicity, race, or BMI in patients with B-ALL. Moreover, among patients with B-ALL, overall survival was similar between demographic groups. These results differ from outcomes observed for Hispanic patients and patients with obesity in previous studies with standard chemotherapy. A secondary analysis of 794 patients, including 150 Hispanic patients, from a phase 3 clinical trial for B-ALL in children at Dana-Farber demonstrated inferior overall survival in Hispanic patients.44 

With respect to BMI, we similarly demonstrate comparable remission rates and survival outcomes between obese vs nonobese patients. Overall, this suggests CAR T-cell therapy may be uniquely situated to provide substantial benefit among high-risk patient populations who are more likely to be chemotherapy resistant, highlighting the need to improve access to CAR T-cell therapy and clinical trials in these groups. In this regard, we unexpectedly identified that Hispanic patients and those with obesity were significantly less likely to have received prior HSCT, suggesting either issues with access to HSCT or having chemotherapy-resistant disease that precluded HSCT–warranting further study.

Despite similar efficacy across demographic groups, our study found a consistent association between Hispanic ethnicity and severe CRS. Although validated models to predict CRS severity following CAR therapy are lacking, 2 strong predictors include baseline bone marrow disease burden in B-ALL and CAR T-cell dose.31 Neither accounted for the trend found in our study. Although high disease burden was associated with grade ≥3 CRS, multivariable analysis, including disease burden, age, and Hispanic ethnicity, found all three to be independently associated with severe CRS. Despite changes in CRS management since the debut of CAR T-cell therapy, we did not observe a consistent trend of CRS severity by treatment year in our B-ALL cohort.

Importantly, other factors not evaluated in this study, such as lymphodepletion regimens and CAR manufacturing nuances,45 have also been implicated with CRS severity, and therefore, additional work is required to elucidate whether these factors might impact the toxicity disparities we found. Notably, extremes of BMI have been associated with greater toxicity with conventional leukemia therapy,46 and although our study failed to show an association between BMI and CRS severity in general, we did find Hispanic patients who were overweight or obese were at the highest risk for severe CRS in our cohort. The basis for this disparity is unclear and warrants further investigation, but previous work in the health disparities space points to multifactorial causes, including differences in cancer biology and preexisting comorbidities in addition to structural barriers to equitable care.47 How this impacts dosing considerations in obese patients receiving CAR T-cells is an additional question that should be more broadly studied. Similarly, our finding that adult patients with B-ALL were at higher risk for severe CRS compared with pediatric patients with B-ALL independent of ethnicity and disease burden warrants further investigation with larger studies.

The incidence of neurotoxicity was comparable to the rate of severe CRS in our study. However, we found no significant associations between ethnicity or BMI and neurotoxicity. The basis for higher incidence of neurotoxicity in male compared with female patients in our study is unknown, although the CI for OR estimate is wide because of the sample size of our study. Previous work has identified common predictors, such as baseline disease burden in B-ALL, for CRS and neurotoxicity.31 Interestingly, baseline disease burden was not significantly associated with neurotoxicity on univariate analysis in our B-ALL cohort, suggesting other drivers of neurotoxicity may be present, but also aligning with the overall lower neurotoxicity rates seen across our phase 1 CAR T-cell trials for B-ALL.48,49 Among these drivers may be neurologic comorbidities present prior to receipt of CAR T-cell therapy. Ongoing work by our group and others has aimed to better understand the determinants of neurotoxicity.

Our study has several limitations, including those inherent to the retrospective nature of this analysis. Patients with B-ALL in our cohort were heavily pretreated before receiving CAR T-cell therapy, with a large percentage of patients having received CAR T-cell therapy or HSCT prior to enrollment in our trials, although CAR T-cell reinfusions (=infusion of the same product) were specifically excluded from this analysis for this reason.50 Previous studies examining the association of obesity with cancer outcomes have typically used BMI at diagnosis, but this was not feasible in our study given that the majority of patients were diagnosed at other institutions many years prior to enrollment in our clinical trials. Although obesity of varying classes may be of interest within the field,51 our analysis is limited by insufficient patients to adequately examine differences between these patient populations, and prospective evaluations may be more definitive.

Socioeconomic status, particularly with respect to family and neighborhood, is also known to significantly contribute to disparities in cancer outcomes,52 but we were unable to adequately account for this due to limited documentation patient insurance status. These factors may represent significant confounders we were unable to control for. The relatively small number of subjects in our study, particularly with respect to representation of Black patients, limits our ability to perform more comprehensive demographic analyses and reduces our power to detect smaller but clinically meaningful differences in efficacy outcomes across subgroups. As patients referred to our institution had multiply refractory disease, overall survival is likely not reflective of outcomes in patients who are treated earlier in their disease course, which is particularly relevant as CAR T cells move earlier into treatment paradigms. Last, although our results highlight the potential of CAR T-cell therapy to improve outcomes in populations who have worse chemotherapy outcomes, it is important to recognize combating cancer health disparities will require a multifaceted approach that incorporates not only better access to emerging therapies such as CAR T cells but also improved prevention and relapse mitigation strategies.

In conclusion, based on this retrospective analysis across 5 phase 1 clinical trials for both adult and pediatric patients with 3 hematologic malignancies treated with CAR T-cell therapy, we show that Hispanic patients are more likely to experience severe CRS after CAR T-cell therapy, even after adjusting for baseline B-ALL disease burden and age. Future larger studies are needed to both validate these findings and further characterize the factors underlying differential toxicity among demographic groups, with the ultimate goal of implementing mitigation strategies that reduce toxicity while maintaining efficacy. Despite this discrepancy in toxicity, efficacy across racial/ethnic groups and BMI classes was comparable. Given that Hispanic patients and patients with obesity are more likely to experience chemotherapy-resistant or refractory disease, our findings suggest that CAR T-cell therapy may provide substantial benefit to these high-risk groups. Thus, efforts to identify barriers in representation of racial/ethnic minorities and other high-risk groups in clinical trials in general, and CAR T-cell therapy trials in particular, should be prioritized.

The authors gratefully acknowledge the study participants and their families, referring medical care teams, the faculty and staff of the NIH Clinical Center who provide their expertise in the management of the study participants, patient care coordinators, and the data managers, Showri Kakumanu and Ekaterina Nikitina, involved with this work on the primary treatment trials. The authors additionally acknowledge Brian Hill (Cleveland Clinic), Amy Nowacki (Cleveland Clinic), and Anupam Verma (NCI) for their critical input into this analysis and the Pediatric Oncology Branch Diversity Working Group for their ongoing efforts in addressing concerns related to diversity and enrollment. This work was supported in part by the Intramural Research Program of the National Institutes of Health (NIH), National Cancer Institute, Center for Cancer Research, and the Warren Grant Magnuson Clinical Center.

This project has been funded in whole or in part with federal funds from the National Institutes of Health, National Cancer Institute grant ZIA BC 011823 (N.N.S.). Research support was also provided by the NIH Medical Research Scholars Program, a public-private partnership supported jointly by the NIH and contributions to the Foundation for the NIH from the American Association for Dental Research and the Colgate-Palmolive Company (A.J.F.) and The Children’s Cancer Foundation (H.S.).

The content of this publication does not necessarily reflect the views of policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the US Government.

Contribution: A.J.F., P.B., and N.N.S. designed the study and performed primary data analysis; A.J.F. and N.N.S. wrote the first version of the manuscript; J.A.L., P.B., and B.Y. provided critical input for the data analysis; S.M.S. conducted statistical analysis and provided critical input on select sections within the manuscript; J.A.L., T.F., L.L., C.L.M., D.W.L., T.J.F., H.S., J.B., B.Y., L.M., J.K., and N.N.S. provided patient care, oversight of the clinical trials for which the subjects were enrolled on, and primary data that contributed to this analysis. No nonauthor wrote the first draft or any part of the paper. All authors contributed to the review of the final manuscript and have agreed to be coauthors.

Conflict-of-interest disclosure: D.W.L. consults for Harpoon Therapeutics, serves on external advisory boards for BMS/Juno Therapeutics and Amgen, and his institution (UVA) receives clinical trial support from Gilead/Kite Pharma. C.L.M. consults for Lyell, Syncopation, Nektar, Bristol Myers Squibb, GlaxoSmithKline, Immatics, Neoimmune Tech, Apricity, Mammoth, and Ensoma, holds equity in Lyell, Syncopation, Apricity, Mammoth, and Ensoma, and receives research funding from Lyell. T.J.F. is an employee of Sana Biotechnology. The remaining authors declare no competing financial interests.

Correspondence: Nirali N. Shah, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; e-mail: Nirali.Shah@nih.gov.

1.
Fesnak
AD
,
June
CH
,
Levine
BL
.
Engineered T cells: the promise and challenges of cancer immunotherapy
.
Nat Rev Cancer
.
2016
;
16
(
9
):
566
-
581
.
2.
Maude
SL
,
Laetsch
TW
,
Buechner
J
, et al
.
Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia
.
N Engl J Med
.
2018
;
378
(
5
):
439
-
448
.
3.
Locke
FL
,
Miklos
DB
,
Jacobson
CA
, et al;
All ZUMA-7 Investigators and Contributing Kite Members
.
Axicabtagene ciloleucel as second-line therapy for large B-cell lymphoma
.
N Engl J Med
.
2022
;
386
(
7
):
640
-
654
.
4.
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
.
5.
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
(
41
):
1
-
20
.
6.
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
.
2021
;
398
(
10297
):
314
-
324
.
7.
Al Hadidi
S
,
Schinke
C
,
Thanendrarajan
S
,
Zangari
M
,
van Rhee
F
.
Enrollment of Black participants in pivotal clinical trials supporting US Food and Drug Administration approval of chimeric antigen receptor-T cell therapy for hematological malignant neoplasms
.
JAMA Netw Open
.
2022
;
5
(
4
):
e228161
.
8.
Ahmed
N
,
Shahzad
M
,
Shippey
E
, et al
.
Socioeconomic and racial disparity in chimeric antigen receptor T cell therapy access
.
Transplant Cell Ther
.
2022
;
28
(
7
):
358
-
364
.
9.
Shoag
JM
,
Barredo
JC
,
Lossos
IS
,
Pinheiro
PS
.
Acute lymphoblastic leukemia mortality in Hispanic Americans
.
Leuk Lymphoma
.
2020
;
61
(
11
):
2674
-
2681
.
10.
Printz
C
.
Neurotoxicity more likely in Hispanic children treated for acute lymphoblastic leukemia
.
Cancer
.
2019
;
125
(
4
):
494
-
495
.
11.
Savage
B
,
Cole
PD
,
Lin
H
,
Thomas-Hawkins
C
.
Hispanic children hospitalized with acute lymphoblastic leukemia are at increased risk of pancreatitis
.
Cancer Nurs
.
2021
;
44
(
6
):
509
-
515
.
12.
Eche
IJ
,
Aronowitz
T
.
A literature review of racial disparities in overall survival of black children with acute lymphoblastic leukemia compared with white children with acute lymphoblastic leukemia
.
J Pediatr Oncol Nurs
.
2020
;
37
(
3
):
180
-
194
.
13.
Sasaki
K
,
Jabbour
E
,
Short
NJ
, et al
.
Acute lymphoblastic leukemia: a population-based study of outcome in the United States based on the surveillance, epidemiology, and end results (SEER) database, 1980-2017
. [published correction appears in Am J Hematol. 2021;96(10):1344].
Am J Hematol
.
2021
;
96
(
6
):
650
-
658
.
14.
Raca
G
,
Abdel-Azim
H
,
Yue
F
, et al
.
Increased incidence of IKZF1 deletions and IGH-CRLF2 translocations in B-ALL of Hispanic/Latino children—a novel health disparity
.
Leukemia
.
2021
;
35
(
8
):
2399
-
2402
.
15.
Nieto
MJ
,
Li
Z
,
Rehman
H
,
Saif
MW
.
Lower 24-month relative survival among black patients with non-Hodgkin’s lymphoma: an analysis of the SEER data 1997-2015
.
J Clin Haematol
.
2021
;
2
(
1
):
5
-
13
.
16.
Derman
BA
,
Jasielec
J
,
Langerman
SS
,
Zhang
W
,
Jakubowiak
AJ
,
Chiu
BC
.
Racial differences in treatment and outcomes in multiple myeloma: a multiple myeloma research foundation analysis
.
Blood Cancer J
.
2020
;
10
(
8
):
80
.
17.
Ghosh
T
,
Richardson
M
,
Gordon
PM
,
Ryder
JR
,
Spector
LG
,
Turcotte
LM
.
Body mass index associated with childhood and adolescent high-risk B-cell acute lymphoblastic leukemia risk: a Children’s Oncology Group report
.
Cancer Med
.
2020
;
9
(
18
):
6825
-
6835
.
18.
Orgel
E
,
Framson
C
,
Buxton
R
, et al
.
Caloric and nutrient restriction to augment chemotherapy efficacy for acute lymphoblastic leukemia: the IDEAL trial
.
Blood Adv
.
2021
;
5
(
7
):
1853
-
1861
.
19.
Mittelman
SD
,
Kim
J
,
Raca
G
,
Li
G
,
Oberley
MJ
,
Orgel
E
.
Increased prevalence of CRLF2 rearrangements in obesity-associated acute lymphoblastic leukemia
.
Blood
.
2021
;
138
(
2
):
199
-
202
.
20.
Butturini
AM
,
Dorey
FJ
,
Lange
BJ
, et al
.
Obesity and outcome in pediatric acute lymphoblastic leukemia
.
J Clin Oncol
.
2007
;
25
(
15
):
2063
-
2069
.
21.
Parikh
R
,
Tariq
SM
,
Marinac
CR
,
Shah
UA
.
A comprehensive review of the impact of obesity on plasma cell disorders
.
Leukemia
.
2022
;
36
(
2
):
301
-
314
.
22.
Leo
QJ
,
Ollberding
NJ
,
Wilkens
LR
, et al
.
Obesity and non-Hodgkin lymphoma survival in an ethnically diverse population: the Multiethnic Cohort study
.
Cancer Causes Control
.
2014
;
25
(
11
):
1449
-
1459
.
23.
Chen
H
,
Wang
D
,
Zhong
Q
,
Tao
Y
,
Zhou
Y
,
Shi
Y
.
Pretreatment body mass index and clinical outcomes in cancer patients following immune checkpoint inhibitors: a systematic review and meta-analysis
.
Cancer Immunol Immunother
.
2020
;
69
(
12
):
2413
-
2424
.
24.
Cortellini
A
,
Ricciuti
B
,
Tiseo
M
, et al
.
Baseline BMI and BMI variation during first line pembrolizumab in NSCLC patients with a PD-L1 expression ≥ 50%: a multicenter study with external validation
.
J Immunother Cancer
.
2020
;
8
(
2
):
e001403
.
25.
Woodall
MJ
,
Neumann
S
,
Campbell
K
,
Pattison
ST
,
Young
SL
.
The effects of obesity on anti-cancer immunity and cancer immunotherapy
.
Cancers (Basel)
.
2020
;
12
(
5
):
1230
.
26.
Christina Baggott
MK
,
Snehit
Prabhu
,
Holly L
Pacenta
,
Christine L
Phillips
,
Jenna
Rossoff
,
Heather E
Stefanski
,
Julie-An
Talano
,
Amy
Moskop
,
Karen
Chao
,
Steven
Margossian
,
Michael R
Verneris
,
Douglas
Myers
,
Nicole
Karras
,
Patrick A
Brown
,
Muna
Qayed
,
Michelle
Hermiston
,
M
Christa Krupski
,
Prakash
Satwani
,
Amy K
Keating
,
Rachel
Wilcox
,
Cara A
Rabik
,
Vanessa A
Fabrizio
,
Vasant
Chinnabhandar
,
Yasemin
Goksenin
,
Crystal
Mackall
,
Smita
Bhatia
,
Kevin J
Curran
,
Theodore W
Laetsch
,
Liora Michal
Schultz
. Inferior outcomes among black patients with childhood acute lymphoblastic leukemia following tisagenlecleucel.
Transplantation & Cellular Therapy Meetings of ASTCT and CIBMTR
;
2021
.
27.
Dos Santos
DMC
,
Rejeski
K
,
Winkelmann
M
, et al
.
Increased visceral fat distribution and body composition impact cytokine release syndrome onset and severity after CD19 CAR-T in advanced B-cell malignancies
.
Haematologica
.
2022
;
107
(
9
):
2096
-
2107
.
28.
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
.
29.
Cheson
BD
,
Fisher
RI
,
Barrington
SF
, et al;
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
.
30.
Durie
BG
,
Harousseau
JL
,
Miguel
JS
, et al;
International Myeloma Working Group
.
International uniform response criteria for multiple myeloma
. [published corrections appear in Leukemia. 2006;20(12):2220; Leukemia. 2007;21(5):1134].
Leukemia
.
2006
;
20
(
9
):
1467
-
1473
.
31.
Sheth
VS
,
Gauthier
J
.
Taming the beast: CRS and ICANS after CAR T-cell therapy for ALL
.
Bone Marrow Transplant
.
2021
;
56
(
3
):
552
-
566
.
32.
Frey
N
,
Porter
D
.
Cytokine release syndrome with chimeric antigen receptor T cell therapy
.
Biol Blood Marrow Transplant
.
2019
;
25
(
4
):
e123
-
e127
.
33.
Shalabi
H
,
Qin
H
,
Su
A
, et al
.
CD19/22 CAR T cells in children and young adults with B-ALL: phase 1 results and development of a novel bicistronic CAR
.
Blood
.
2022
;
140
(
5
):
451
-
463
.
34.
Davis
TC
,
Arnold
CL
,
Mills
G
,
Miele
L
.
A qualitative study exploring barriers and facilitators of enrolling underrepresented populations in clinical trials and biobanking
.
Front Cell Dev Biol
.
2019
;
7
:
74
.
35.
Bishop
MR
,
Dickinson
M
,
Purtill
D
, et al
.
Second-line tisagenlecleucel or standard care in aggressive B-cell lymphoma
.
N Engl J Med
.
2022
;
386
(
7
):
629
-
639
.
36.
Awidi
M
,
Al Hadidi
S
.
Participation of Black Americans in cancer clinical trials: current challenges and proposed solutions
.
JCO Oncol Pract
.
2021
;
17
(
5
):
265
-
271
.
37.
Camidge
DR
,
Park
H
,
Smoyer
KE
, et al
.
Race and ethnicity representation in clinical trials: findings from a literature review of phase I oncology trials
.
Future Oncol
.
2021
;
17
(
24
):
3271
-
3280
.
38.
Wudhikarn
K
,
Bansal
R
,
Khurana
A
, et al
.
The impact of obesity and body weight on the outcome of patients with relapsed/refractory large B-cell lymphoma treated with axicabtagene ciloleucel
.
Blood Cancer J
.
2021
;
11
(
7
):
124
.
39.
Shah
NN
,
Highfill
SL
,
Shalabi
H
, et al
.
CD4/CD8 T-cell selection affects chimeric antigen receptor (CAR) T-cell potency and toxicity: updated results from a phase I anti-CD22 CAR T-cell trial
.
J Clin Oncol
.
2020
;
38
(
17
):
1938
-
1950
.
40.
Shah
NN
,
Lee
DW
,
Yates
B
, et al
.
Long-term follow-up of CD19-CAR T-cell therapy in children and young adults with B-ALL
.
J Clin Oncol
.
2021
;
39
(
15
):
1650
-
1659
.
41.
Brudno
JN
,
Lam
N
,
Vanasse
D
, et al
.
Safety and feasibility of anti-CD19 CAR T cells with fully human binding domains in patients with B-cell lymphoma
. [published correction appears in Nat Med. 2020;26(5):803].
Nat Med
.
2020
;
26
(
2
):
270
-
280
.
42.
Brudno
JN
,
Maric
I
,
Hartman
SD
, et al
.
T cells genetically modified to express an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of poor-prognosis relapsed multiple myeloma
.
J Clin Oncol
.
2018
;
36
(
22
):
2267
-
2280
.
43.
Ali
SA
,
Shi
V
,
Maric
I
, et al
.
T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma
.
Blood
.
2016
;
128
(
13
):
1688
-
1700
.
44.
Kahn
JM
,
Cole
PD
,
Blonquist
TM
, et al
.
An investigation of toxicities and survival in Hispanic children and adolescents with ALL: results from the Dana-Farber Cancer Institute ALL Consortium protocol 05-001
.
Pediatr Blood Cancer
.
2018
;
65
(
3
):
e26871
.
45.
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
.
46.
Orgel
E
,
Sposto
R
,
Malvar
J
, et al
.
Impact on survival and toxicity by duration of weight extremes during treatment for pediatric acute lymphoblastic leukemia: a report from the Children’s Oncology Group
.
J Clin Oncol
.
2014
;
32
(
13
):
1331
-
1337
.
47.
Kirtane
K
,
Lee
SJ
.
Racial and ethnic disparities in hematologic malignancies
.
Blood
.
2017
;
130
(
15
):
1699
-
1705
.
48.
Shalabi
H
,
Martin
S
,
Yates
B
, et al
.
Neurotoxicity following CD19/CD28ζ CAR T-cells in children and young adults with B-cell malignancies
. [published online ahead of print 11 February 2022].
Neuro-oncol
.
2022
:
noac034
.
49.
Shalabi
H
,
Wolters
PL
,
Martin
S
, et al
.
Systematic evaluation of neurotoxicity in children and young adults undergoing CD22 chimeric antigen receptor T-cell therapy
.
J Immunother
.
2018
;
41
(
7
):
350
-
358
.
50.
Myers
RM
,
Devine
K
,
Li
Y
, et al
. Outcomes after reinfusion of CD19-specific chimeric antigen receptor (CAR)-modified T cells in children and young adults with relapsed/refractory B-cell acute lymphoblastic leukemia.
American Society of Hematology Annual Meeting & Exposition: ASH Publications
;
2021
.
51.
Advani
AS
,
Larsen
E
,
Laumann
K
, et al
.
Comparison of CALGB 10403 (Alliance) and COG AALL0232 toxicity results in young adults with acute lymphoblastic leukemia
.
Blood Adv
.
2021
;
5
(
2
):
504
-
512
.
52.
Ward
E
,
Jemal
A
,
Cokkinides
V
, et al
.
Cancer disparities by race/ethnicity and socioeconomic status
.
CA Cancer J Clin
.
2004
;
54
(
2
):
78
-
93
.

Author notes

Presented in abstract form at the 2022 American Society of Pediatric Hematology/Oncology Conference, Pittsburgh, PA, 3 May 2022.

Data are available in a limited capacity by request to the corresponding author: nirali.shah@nih.gov.

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

Supplemental data