• Overweight refractory/relapsed myeloma patients had worse outcomes with anti-BCMA CAR T-cell therapy than normal weight or obese patients.

Abstract

Anti–B-cell maturation antigen (BCMA) chimeric antigen receptor (CAR) T-cell therapy has transformed treatment of relapsed/refractory multiple myeloma (MM), yet host determinants of efficacy remain poorly defined. Although obesity influences immunotherapy outcomes in numerous cancers, the relationship between body mass index (BMI) and CAR T-cell therapy outcomes in MM is unclear. We retrospectively evaluated MM patients who were treated with anti-BCMA CAR T-cell therapy at Massachusetts General Hospital from 2016 to 2023. Cubic spline modeling revealed a nonlinear U-shaped association between BMI and progression-free survival (PFS). Patients who were overweight (BMI, 25-29.9 kg/m2) displayed worse outcomes than those with normal weight (BMI < 25 kg/m2), or those who were obese (BMI ≥ 30.0 kg/m2): 12-month PFS was 28.8% vs 51.9% and 62.6% (P < .001); 12-month overall survival (OS) was 61.4% vs 82.9% and 84.2% (P = .006); complete response rate was 36.4% vs 42.9% and 56.1% (P = .185) in the overweight, normal weight, and obese groups, respectively. Patients who were overweight also had inferior PFS and OS in multivariable models. Cytokine release syndrome and immune effector cell–associated neurotoxicity syndrome incidence and severity did not significantly differ by BMI. These data support a U-shaped relationship between BMI and CAR T-cell outcomes in MM, which should motivate mechanistic studies to identify modifiable biologic factors.

The obesity epidemic is a global health problem that has been linked to the development of numerous malignancies,1 including multiple myeloma (MM).2 Obesity also affects cancer immunotherapy responses:3 obese patients with solid tumors have superior responses to immune checkpoint inhibitors (ICI),4 and patients with B-cell lymphoma with increased visceral fat respond better to anti-CD19 chimeric antigen receptor (CAR) T-cell therapy.5 This poorly understood phenomenon is called the “obesity paradox,”6 and highlights the complex interaction between metabolism, antitumor immunity, and cancer biology.

CAR T-cell therapy targeting B-cell maturation antigen (BCMA) has transformed the treatment of relapsed/refractory MM.7 Idecabtagene vicleucel and ciltacabtagene autoleucel are the 2 current US Food and Drug Administration-approved CAR T-cell therapies in MM. Modifiable host factors, such as metabolic health and obesity, that impact response and relapse after CAR T-cell therapy are an area of active investigation. A report from China evaluating investigational anti-BCMA CAR T-cell constructs found that patients with body mass index (BMI) ≥25 kg/m2 had prolonged progression-free survival (PFS) compared with those with BMI <25 kg/m2.8 However, the impact of BMI on outcomes in a United States cohort has not been described.

Because BMI conflates adiposity and lean mass, its association with clinical outcomes may be complex. We hypothesized that normal weight patients have more favorable tumor biology, whereas obesity may paradoxically confer enhanced immunotherapy sensitivity and CAR T-cell therapy efficacy, as seen in other cancers.6,9,10 Thus, the intermediary overweight state may represent a relatively immunometabolically adverse phenotype without the compensatory benefits seen at either end of the BMI spectrum (Figure 1A). To evaluate this hypothesis, we modeled the relationship between BMI and CAR T-cell therapy outcomes using a restricted cubic spline11—an approach that captures nonlinear associations between continuous variables—to evaluate how BMI influences CAR T-cell therapy outcomes.

Figure 1.

CAR T-cell responses separated by BMI. (A) Hypothetical model for relationship between BMI and CAR T-cell therapy outcomes. (B) A Cox proportional hazards model was used to evaluate the association between BMI and PFS. A cubic spline model with 3 degrees of freedom was applied to assess for the presence of a nonlinear relationship between BMI and PFS. The observed HR across the BMI range with 95% CI is shown. (C) Best objective response after CAR T-cell therapy shown for the normal weight (BMI < 25.0 kg/m2), overweight (BMI 25.0-29.9 kg/m2), and obese (BMI ≥ 30.0 kg/m2) groups. PFS (D) and OS (E) for the normal weight, overweight, and obese groups. CR, complete response; HR, hazard ratio; ORR, overall response rate; PR, partial response; VGPR, very good partial response.

Figure 1.

CAR T-cell responses separated by BMI. (A) Hypothetical model for relationship between BMI and CAR T-cell therapy outcomes. (B) A Cox proportional hazards model was used to evaluate the association between BMI and PFS. A cubic spline model with 3 degrees of freedom was applied to assess for the presence of a nonlinear relationship between BMI and PFS. The observed HR across the BMI range with 95% CI is shown. (C) Best objective response after CAR T-cell therapy shown for the normal weight (BMI < 25.0 kg/m2), overweight (BMI 25.0-29.9 kg/m2), and obese (BMI ≥ 30.0 kg/m2) groups. PFS (D) and OS (E) for the normal weight, overweight, and obese groups. CR, complete response; HR, hazard ratio; ORR, overall response rate; PR, partial response; VGPR, very good partial response.

Close modal

We retrospectively studied patients treated with anti-BCMA CAR T-cell therapy for relapsed/refractory MM at Massachusetts General Hospital from 2016 to 2023. The study was approved by our institutional review board, and was conducted in accordance with the Declaration of Helsinki. Baseline characteristics including BMI and inflammatory serologic markers were recorded on the start date of lymphodepletion (LD). The Kruskal-Wallis test and Fisher exact test were used to compare continuous and categorical variables, respectively. Responses were defined according to International Myeloma Working Group criteria.12 High-risk cytogenetics were defined as del(17p), t(4;14), or t(14;16), by fluorescence in situ hybridization, per International Myeloma Working Group criteria.12 PFS was defined as the time from CAR T-cell infusion to either disease progression or death. Overall survival (OS) was defined as the time from CAR T-cell infusion to death. Cytokine release syndrome (CRS) and immune effector cell–associated neurotoxicity syndrome (ICANS) were graded according to American Society for Transplantation and Cellular Therapy (ASTCT) guidelines.13 A Cox proportional hazards regression model incorporating a restricted cubic spline with 3 degrees of freedom11 was used to evaluate the relationship between BMI and PFS. PFS and OS probabilities were estimated using the Kaplan-Meier method, and compared between subgroups with log-rank tests. Univariable and multivariate Cox proportional hazards models were developed for PFS and OS; hazard ratios (HR) were reported with 95% confidence intervals (CI). Statistically significant predictors (P < .05) in univariable models were included in multivariable models. BMI and baseline inflammatory laboratory value associations were modeled using linear regression. Analyses were performed with R 4.5.1.

Among 134 patients with MM (56.7% male) treated with anti-BCMA CAR T-cell therapy (idecabtagene vicleucel 51.5%, ciltacabtagene autoleucel 23.9%, investigational 24.6%), the median age was 67 years (39-85 years), median number of prior therapy lines was 4 (1-16), and 53% had prior autologous hematopoietic cell transplant (AHCT). At CAR T-cell infusion, 52.2% had extramedullary disease and 38.1% had high-risk cytogenetics. Median apheresis-to-infusion time was 41 days, and 73.9% received bridging therapy. Median BMI was 26.7 kg/m2 (17.9-55.2 kg/m2). The median follow-up time from CAR T-cell infusion was 19.9 months (95% CI, 16.6-29.8).

A Cox model with restricted cubic spline revealed a nonlinear BMI-PFS relationship with increased HR in the mid-range (Figure 1B). Patients were grouped as normal weight (n = 49, 36.6%), overweight (n = 44, 32.8%), and obese (n = 41, 30.6%). Baseline characteristics were generally balanced between groups (Table 1), with more males in the overweight and obese groups (P < .001). In the overweight group, more patients received prior AHCT (P = .003), and had lower pre-LD white blood cell (P = .007) and platelet counts (P = .040). Diabetes was more prevalent in the obese group (26.8% vs 6.1% and 13.6%; P = .24); hypertension/hyperlipidemia and pre-LD C-reactive protein (CRP) also positively trended with BMI.

Table 1.

Baseline characteristics stratified by BMI group

VariableNormal weightOverweightObeseP value
BMI < 25 kg/m2BMI, 25-29.9 kg/m2BMI ≥ 30 kg/m2
N = 49N = 44N = 41
BMI (kg/m2)     
Median [min-max] 22.7 [17.9-24.9] 27.3 [25.0-29.9] 34.9 [30.2-55.2] – 
Age at CAR T-cell therapy (y)     
Median [min, max] 68 [50-78] 68 [39-85] 65 [46-83] .203 
Sex     
Male 17 (34.7%) 30 (68.2%) 29 (70.7%) <.001 
Race     
White 39 (79.6%) 34 (77.3%) 36 (87.8%) .510 
Black 3 (6.1%) 4 (9.1%) 2 (4.9%)  
Asian 5 (10.2%) 1 (2.3%) 1 (2.4%)  
Hispanic/Latino 2 (4.1%) 4 (9.1%) 1 (2.4%)  
Other 0 (0%) 1 (2.3%) 1 (2.4%)  
Lines of therapy prior to     
CAR T-cell therapy     
Median [min-max] 4 [1-10] 5 [1-16] 4 [1-11] .135 
MM subtype     
IgG kappa 19 (38.8%) 14 (31.8%) 11 (26.8%) .972 
IgG lambda 8 (16.3%) 7 (15.9%) 8 (19.5%)  
IgA kappa 5 (10.2%) 5 (11.4%) 4 (9.8%)  
IgA lambda 2 (4.1%) 5 (11.4%) 4 (9.8%)  
Kappa light chain 8 (16.3%) 6 (13.6%) 6 (14.6%)  
Lambda light chain 7 (14.3%) 7 (15.9%) 7 (17.1%)  
Nonsecretory 0 (0%) 0 (0%) 1 (2.4%)  
Extramedullary disease at CAR T-cell therapy     
Yes 24 (49.0%) 27 (61.4%) 19 (46.3%) .546 
No 17 (34.7%) 10 (22.7%) 16 (39.0%)  
Unknown 8 (16.3%) 7 (15.9%) 6 (14.6%)  
High-risk cytogenetics     
Yes 17 (34.7%) 21 (47.7%) 13 (31.7%) .181 
No 30 (61.2%) 19 (43.2%) 26 (63.4%)  
Not available 2 (4.1%) 4 (9.1%) 2 (4.9%)  
Prior AHCT     
Yes 19 (38.8%) 32 (72.7%) 20 (48.8%) .003 
ECOG performance status at CAR T-cell therapy     
20 (40.8%) 17 (38.6%) 8 (19.5%) .190 
24 (49.0%) 24 (54.5%) 29 (70.7%)  
≥2 5 (10.2%) 3 (6.8%) 4 (9.8%)  
Hypertension     
Yes 27 (55.1%) 25 (56.8%) 30 (73.2%) .177 
Hyperlipidemia     
Yes 18 (36.7%) 18 (40.9%) 22 (53.7%) .253 
Diabetes mellitus     
Yes 3 (6.1%) 6 (13.6%) 11 (26.8%) .024 
Anti-BCMA CAR T-cell construct     
Idecabtagene vicleucel 20 (40.8%) 28 (63.6%) 21 (51.2%) .266 
Ciltacabtagene autoleucel 14 (28.6%) 7 (15.9%) 11 (26.8%)  
Investigational 15 (30.6%) 9 (20.5%) 9 (22.0%)  
Time from apheresis to CAR T-cell infusion (d)     
Median [min-max] 42 [29-82] 40 [21-74] 41 [30-92] .231 
Bridging therapy prior to CAR T-cell therapy     
Yes 37 (75.5%) 32 (72.7%) 30 (73.2%) .939 
White blood cells (×109/L)      
Median [min-max] 3.86 [1.32-20.0] 3.21 [1.79-6.78] 3.91 [1.50-12.1] .007 
Neutrophils (×109/L)      
Median [min-max] 2.47 [0.89-16.80] 1.92 [0.59-5.95] 2.71 [0.80-9.82] .016 
Hemoglobin (g/dL)      
Median [min-max] 9.8 [5.7-12.4] 10.0 [7.4-14.8] 10.6 [7.0-14.8] .108 
Platelets (×109/L)      
Median [min-max] 160 [9-381] 113 [4-286] 146 [20-387] .040 
Ferritin (ng/mL)      
Median [min-max] 200 [14-4700] 376 [20-8420] 266 [12-4460] .445 
CRP (mg/L)      
Median [min-max] 3.2 [0.2-127] 5.0 [0.2-841] 7.4 [0.7-289] .067 
LDH (U/L)      
Median [min-max] 189 [122-879] 184 [93-648] 186 [111-1700] .523 
VariableNormal weightOverweightObeseP value
BMI < 25 kg/m2BMI, 25-29.9 kg/m2BMI ≥ 30 kg/m2
N = 49N = 44N = 41
BMI (kg/m2)     
Median [min-max] 22.7 [17.9-24.9] 27.3 [25.0-29.9] 34.9 [30.2-55.2] – 
Age at CAR T-cell therapy (y)     
Median [min, max] 68 [50-78] 68 [39-85] 65 [46-83] .203 
Sex     
Male 17 (34.7%) 30 (68.2%) 29 (70.7%) <.001 
Race     
White 39 (79.6%) 34 (77.3%) 36 (87.8%) .510 
Black 3 (6.1%) 4 (9.1%) 2 (4.9%)  
Asian 5 (10.2%) 1 (2.3%) 1 (2.4%)  
Hispanic/Latino 2 (4.1%) 4 (9.1%) 1 (2.4%)  
Other 0 (0%) 1 (2.3%) 1 (2.4%)  
Lines of therapy prior to     
CAR T-cell therapy     
Median [min-max] 4 [1-10] 5 [1-16] 4 [1-11] .135 
MM subtype     
IgG kappa 19 (38.8%) 14 (31.8%) 11 (26.8%) .972 
IgG lambda 8 (16.3%) 7 (15.9%) 8 (19.5%)  
IgA kappa 5 (10.2%) 5 (11.4%) 4 (9.8%)  
IgA lambda 2 (4.1%) 5 (11.4%) 4 (9.8%)  
Kappa light chain 8 (16.3%) 6 (13.6%) 6 (14.6%)  
Lambda light chain 7 (14.3%) 7 (15.9%) 7 (17.1%)  
Nonsecretory 0 (0%) 0 (0%) 1 (2.4%)  
Extramedullary disease at CAR T-cell therapy     
Yes 24 (49.0%) 27 (61.4%) 19 (46.3%) .546 
No 17 (34.7%) 10 (22.7%) 16 (39.0%)  
Unknown 8 (16.3%) 7 (15.9%) 6 (14.6%)  
High-risk cytogenetics     
Yes 17 (34.7%) 21 (47.7%) 13 (31.7%) .181 
No 30 (61.2%) 19 (43.2%) 26 (63.4%)  
Not available 2 (4.1%) 4 (9.1%) 2 (4.9%)  
Prior AHCT     
Yes 19 (38.8%) 32 (72.7%) 20 (48.8%) .003 
ECOG performance status at CAR T-cell therapy     
20 (40.8%) 17 (38.6%) 8 (19.5%) .190 
24 (49.0%) 24 (54.5%) 29 (70.7%)  
≥2 5 (10.2%) 3 (6.8%) 4 (9.8%)  
Hypertension     
Yes 27 (55.1%) 25 (56.8%) 30 (73.2%) .177 
Hyperlipidemia     
Yes 18 (36.7%) 18 (40.9%) 22 (53.7%) .253 
Diabetes mellitus     
Yes 3 (6.1%) 6 (13.6%) 11 (26.8%) .024 
Anti-BCMA CAR T-cell construct     
Idecabtagene vicleucel 20 (40.8%) 28 (63.6%) 21 (51.2%) .266 
Ciltacabtagene autoleucel 14 (28.6%) 7 (15.9%) 11 (26.8%)  
Investigational 15 (30.6%) 9 (20.5%) 9 (22.0%)  
Time from apheresis to CAR T-cell infusion (d)     
Median [min-max] 42 [29-82] 40 [21-74] 41 [30-92] .231 
Bridging therapy prior to CAR T-cell therapy     
Yes 37 (75.5%) 32 (72.7%) 30 (73.2%) .939 
White blood cells (×109/L)      
Median [min-max] 3.86 [1.32-20.0] 3.21 [1.79-6.78] 3.91 [1.50-12.1] .007 
Neutrophils (×109/L)      
Median [min-max] 2.47 [0.89-16.80] 1.92 [0.59-5.95] 2.71 [0.80-9.82] .016 
Hemoglobin (g/dL)      
Median [min-max] 9.8 [5.7-12.4] 10.0 [7.4-14.8] 10.6 [7.0-14.8] .108 
Platelets (×109/L)      
Median [min-max] 160 [9-381] 113 [4-286] 146 [20-387] .040 
Ferritin (ng/mL)      
Median [min-max] 200 [14-4700] 376 [20-8420] 266 [12-4460] .445 
CRP (mg/L)      
Median [min-max] 3.2 [0.2-127] 5.0 [0.2-841] 7.4 [0.7-289] .067 
LDH (U/L)      
Median [min-max] 189 [122-879] 184 [93-648] 186 [111-1700] .523 

ECOG, Eastern Cooperative Oncology Group; Ig, immunoglobulin; LDH, lactate dehydrogenase; Max, maximum; Min, minimum.

On start date of LD.

Best overall response rates were 87.8%, 75.0%, and 87.8% (P = .126), and complete response rates were 42.9%, 36.4%, and 56.1% (P = .185) in the normal weight, overweight, and obese groups, respectively (Figure 1C). More patients in the overweight group failed to achieve at least a partial response compared with the combined normal weight and obese groups (25.0% vs 12.2%, P = .040). For the entire cohort, 12-month PFS and OS were 47.8% (95% CI, 39.6-57.8) and 75.8% (95% CI, 68.4-84.0), respectively. Stratified by BMI, outcomes were worst in the overweight group (Figure 1D-E): 12-month PFS was 28.8% vs 51.9% (normal weight) and 62.6% (obese), P < .001; 12-month OS was 61.4% vs 82.9% (normal weight) and 84.2% (obese), P = .006. In univariable Cox models (supplemental Tables 1 and 2), overweight BMI predicted inferior PFS (HR, 2.35 [95% CI, 1.5-3.67]; P < .001) and OS (HR, 2.38 [95% CI, 1.32-4.28]; P = .004). In multivariable models adjusted for significant univariable covariates (eg, sex, number of prior lines of therapy, prior AHCT, CAR T-cell construct, high-risk cytogenetics, pre-LD hemoglobin, platelets, and CRP), overweight BMI remained independently associated with worse PFS (HR, 1.69 [95% CI, 1.03-2.77]; P = .038) and worse OS (HR, 2.33 [95% CI, 1.08-5.032]; P = .031; supplemental Tables 3 and 4). Kaplan-Meier analyses of PFS stratified by extramedullary disease, prior AHCT, prior therapy-line burden, and CAR T-cell construct consistently showed inferior outcomes in the overweight group (Figure 2A-I).

Figure 2.

PFS for BMI groups separated by disease status. PFS for the normal weight, overweight, and obese groups separated by patients with extramedullary disease at time of CAR T-cell therapy (A), without extramedullary disease at time of CAR T-cell therapy (B), with a history of AHCT prior to CAR T-cell therapy (C), without a history of AHCT prior to CAR T-cell therapy (D), with a history of ≥5 lines of therapy prior to CAR T-cell therapy (E), with a history of ≤4 lines of therapy prior to CAR T-cell therapy (F), who received idecabtagene vicleucel (G), who received ciltacabtagene autoleucel (H), or who received an investigational BCMA-targeted CAR T-cell product (I). P value (log-rank) is shown.

Figure 2.

PFS for BMI groups separated by disease status. PFS for the normal weight, overweight, and obese groups separated by patients with extramedullary disease at time of CAR T-cell therapy (A), without extramedullary disease at time of CAR T-cell therapy (B), with a history of AHCT prior to CAR T-cell therapy (C), without a history of AHCT prior to CAR T-cell therapy (D), with a history of ≥5 lines of therapy prior to CAR T-cell therapy (E), with a history of ≤4 lines of therapy prior to CAR T-cell therapy (F), who received idecabtagene vicleucel (G), who received ciltacabtagene autoleucel (H), or who received an investigational BCMA-targeted CAR T-cell product (I). P value (log-rank) is shown.

Close modal

In the entire cohort, any-grade CRS occurred in 89.6% of patients, with grade ≥2 in 29.8% and grade ≥3 in 3.7%; any-grade ICANS occurred in 23.1%, with grade ≥2 in 16.4% and grade ≥3 in 6.7%. The 10-day cumulative incidence of grade ≥2 CRS trended higher in normal weight (36.7%) vs overweight (18.2%) and obese (26.8%) groups (P = .15; supplemental Figure 1A); grade ≥2 ICANS did not significantly differ (supplemental Figure 1B). Given known obesity-inflammation links, we modeled the relationship of BMI and pre-LD ferritin, CRP, and lactate dehydrogenase, but found no significant linear associations (ferritin P = .575, R2 = 0.002; CRP P = .567, R2 = 0.003; lactate dehydrogenase P = .316, R2 = 0.007; supplemental Figure 2A-C).

Collectively, our data support a U-shaped BMI-outcome relationship after anti-BCMA CAR T-cell therapy in MM, with worse responses and survival in patients who were overweight compared with those with normal weight and those with obesity. A potential unifying hypothetical framework is an immunometabolic valley resulting in lower CAR T-cell efficacy in the overweight BMI range, with favorable tumor biology on one end of the BMI spectrum (normal weight) and improved CAR T-cell functionality or sensitivity on the other (obese; Figure 1A). Adipose tissue promotes plasma cell survival/proliferation via interleukin-6, interleukin-1b, tumor necrosis factor-α, and adipokines,1 and obesity is a risk factor for MM progression.1 In the longitudinal CoMMpass study, patients with obesity had significantly shorter PFS and OS compared with normal weight patients.14 Increased visceral fat was also associated with worse treatment response in a German study,15 thus obesity may negatively impact therapy responsiveness.16 Conversely, patients with obesity have superior ICI responses in numerous cancers.6 Proposed explanations for this paradox include the obese T-cell exhaustion phenotype, distinct cytokine profiles, and increased tumor immunogenicity due to impaired immunoediting that promote ICI responsiveness.17,18 However, it is unclear if obesity influences CAR T-cell outcomes. Prior work on BMI and anti-CD19 CAR T-cell response in B-cell lymphoma and acute lymphoblastic leukemia have been mixed: 1 study reported improved outcomes with obesity,19 whereas other studies found no association.20,21 Given that CAR T-cell efficacy is the result of myriad factors, including tumor biology, quality of the apheresis T-cell pool, product phenotype, host responses to LD, preinfusion inflammation, postinfusion expansion, immune cell trafficking, and CAR T-cell persistence, our observation that obesity correlates with better outcomes after anti-BCMA CAR T-cell therapy in MM warrants targeted mechanistic study.

Caveats of our study include (1) the single-center design and CAR T-cell product heterogeneity, resulting in possible construct-specific effects, (2) unintentional selection bias of patient referrals that may have enriched the overweight group for unmeasured adverse risk factors, (3) combination of underweight (BMI < 18.5 kg/m2) with normal weight (BMI 18.5-25 kg/m2) groups together as BMI <25 kg/m2, as the underweight cohort was too small to analyze independently, thus the potential effect of cachexia on anti-MM CAR T-cell responses remains unaddressed, and (4) assessment of BMI only at time of LD, without longitudinal trajectories, as preinfusion weight loss has been linked to worse outcomes with anti-CD19 CAR T-cell therapy.22 

Finally, BMI is an imperfect surrogate that may not reflect adipose mass or metabolic health.23 To better delineate adipose-specific effects, studies utilizing comprehensive body composition analysis are warranted.5 Nevertheless, we provide evidence that both normal weight and obese patients with MM have improved outcomes with anti-BCMA CAR T-cell therapy compared with those who were overweight. Further validation and mechanistic research are essential to elucidate the underlying biological processes. This research could uncover novel therapeutic targets or modifiable factors to be leveraged to improve CAR T-cell therapy outcomes in MM.

A.Y. receives research funding from Amgen, Bristol Myers Squibb, GSK, Johnson & Johnson, and Sanofi. N.R. receives funding from the Multiple Myeloma Research Fund Challenge Award, and the Paula and Rodger Riney Foundation.

Contributions: R.C., K.M., D.C., and N.R. conceived the project idea; R.C., K.M., A.J., C.M., S.S.H., A. Barselau, and D.C. were responsible for collecting patient data and constructing a retrospective patient database; K.M. performed the statistical analyses; R.C., K.M., and D.C. wrote the manuscript; A.Y., A. Branagan, B.P., M.F., N.R., and D.C. were the physicians who took care of the patients included in the study and helped provide data for analysis; and A.Y., A. Branagan, B.P., M.F., and N.R. provided feedback on the manuscript.

Conflict-of-interest disclosure: A.Y. is a consultant for AbbVie, Adaptive Biotechnologies, Amgen, Bristol Myers Squibb (BMS), Celgene, GSK, Johnson & Johnson, Karyopharm, Oncopeptides, Pfizer, Prothena, Regeneron, Sanofi, Sebia, and Takeda. M.F. is a consultant for BMS, Kite/Gilead, Johnson & Johnson/Legend, Cytoagents, and SOBI. The remaining authors declare no competing financial interests.

Correspondence: Diana Cirstea, Center for Multiple Myeloma, Yawkey Building Suite 9A, Massachusetts General Hospital, Boston, MA 02114; email: dcirstea@mgh.harvard.edu.

1.
Choa
R
,
Panaroni
C
,
Bhatia
R
,
Raje
N
.
It is worth the weight: obesity and the transition from monoclonal gammopathy of undetermined significance to multiple myeloma
.
Blood Adv
.
2023
;
7
(
18
):
5510
-
5523
.
2.
Chen
J
,
Dalerba
P
,
Terry
MB
,
Yang
W
.
Global obesity epidemic and rising incidence of early-onset cancers
.
J Glob Health
.
2024
;
14
:
04205
.
3.
Dyck
L
,
Lynch
L
.
Diverse effects of obesity on antitumor immunity and immunotherapy
.
Trends Mol Med
.
2023
;
29
(
2
):
112
-
123
.
4.
An
Y
,
Wu
Z
,
Wang
N
, et al
.
Association between body mass index and survival outcomes for cancer patients treated with immune checkpoint inhibitors: a systematic review and meta-analysis
.
J Transl Med
.
2020
;
18
(
1
):
235
.
5.
Rejeski
K
,
Cordas Dos Santos
DM
,
Parker
NH
, et al
.
Influence of adipose tissue distribution, sarcopenia, and nutritional status on clinical outcomes after CD19 CAR T-cell therapy
.
Cancer Immunol Res
.
2023
;
11
(
6
):
707
-
719
.
6.
Alden
SL
,
Charmsaz
S
,
Li
HL
, et al
.
Pan-tumor analysis to investigate the obesity paradox in immune checkpoint blockade
.
J Immunother Cancer
.
2025
;
13
(
1
):
e009734
.
7.
Raje
N
,
Berdeja
J
,
Lin
Y
, et al
.
Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma
.
N Engl J Med
.
2019
;
380
(
18
):
1726
-
1737
.
8.
Cheng
H
,
Sun
Y
,
Zhang
X
, et al
.
Complex association of body mass index and outcomes in patients with relapsed and refractory multiple myeloma treated with CAR-T cell immunotherapy
.
Cytotherapy
.
2024
;
26
(
8
):
832
-
841
.
9.
McQuade
JL
,
Daniel
CR
,
Hess
KR
, et al
.
Association of body-mass index and outcomes in patients with metastatic melanoma treated with targeted therapy, immunotherapy, or chemotherapy: a retrospective, multicohort analysis
.
Lancet Oncol
.
2018
;
19
(
3
):
310
-
322
.
10.
Wang
Z
,
Aguilar
EG
,
Luna
JI
, et al
.
Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade
.
Nat Med
.
2019
;
25
(
1
):
141
-
151
.
11.
Gauthier
J
,
Wu
QV
,
Gooley
TA
.
Cubic splines to model relationships between continuous variables and outcomes: a guide for clinicians
.
Bone Marrow Transplant
.
2020
;
55
(
4
):
675
-
680
.
12.
Kumar
S
,
Paiva
B
,
Anderson
KC
, et al
.
International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma
.
Lancet Oncol
.
2016
;
17
(
8
):
e328
-
e346
.
13.
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
.
14.
Ren
L
,
Xu
T
,
Ren
Y
,
Liu
P
.
The obesity paradox in multiple myeloma: A report from Multiple Myeloma Research Foundation (MMRF) dataset
.
Cancer Med
.
2023
;
12
(
23
):
21400
-
21407
.
15.
GroΔ
JP
,
Nattenmüller
J
,
Hemmer
S
, et al
.
Body fat composition as predictive factor for treatment response in patients with newly diagnosed multiple myeloma - subgroup analysis of the prospective GMMG MM5 trial
.
Oncotarget
.
2017
;
8
(
40
):
68460
-
68471
.
16.
Shah
UA
,
Whiting
K
,
Devlin
S
, et al
.
Extreme body mass index and survival in newly diagnosed multiple myeloma patients
.
Blood Cancer J
.
2023
;
13
(
1
):
13
.
17.
Naik
A
,
Monjazeb
AM
,
Decock
J
.
The obesity paradox in cancer, tumor immunology, and immunotherapy: potential therapeutic implications in triple negative breast cancer
.
Front Immunol
.
2019
;
10
:
1940
.
18.
Farag
KI
,
Makkouk
A
,
Norian
LA
.
Re-evaluating the effects of obesity on cancer immunotherapy outcomes in renal cancer: what do we really know?
.
Front Immunol
.
2021
;
12
:
668494
.
19.
Schultze-Florey
R
,
Sirvent
A
,
Yakoub-Agha
I
, et al
.
High body mass index is associated with favorable outcome in younger patients receiving CD19 CAR T-cell therapy for B-cell lymphoma: a retrospective study from the EBMT Transplant Complications Working Party
.
Blood
.
2023
;
142
(
suppl 1
):
4867
.
20.
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
.
21.
Faruqi
AJ
,
Ligon
JA
,
Borgman
P
, et al
.
The impact of race, ethnicity, and obesity on CAR T-cell therapy outcomes
.
Blood Adv
.
2022
;
6
(
23
):
6040
-
6050
.
22.
Valtis
YK
,
Devlin
S
,
Shouval
R
, et al
.
Cancer cachexia and weight loss before CAR T-cell therapy for lymphoma are independently associated with poor outcomes
.
Blood Adv
.
2025
;
9
(
1
):
151
-
161
.
23.
Wu
Y
,
Li
D
,
Vermund
SH
.
Advantages and limitations of the body mass index (BMI) to assess adult obesity
.
Int J Environ Res Public Health
.
2024
;
21
(
6
):
757
.

Author notes

R.C. and K.M. contributed equally to this work.

The data from this study are available from the corresponding author, Diana Cirstea (dcirstea@mgh.harvard.edu), on request.

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

Supplemental data