Introduction: This study was designed to compare outcomes among patients by race and ethnicity in the post-covalent Bruton tyrosine kinase inhibitor (cBTKi) treatment era. Methods: A nationwide electronic health record (EHR)-derived de-identified database was utilized that included patients diagnosed with CLL from 2013 to 2022 who received systemic therapy for their disease. Use of cBTKi therapy, time to next treatment or death (TTNT-D), and overall survival (OS) were compared by race in unadjusted (Kaplan-Meier method) and adjusted analyses (Cox proportional hazards regression). Results: This study included 4,572 White (71.8%) and 558 Black (8.8%) patients with CLL; 270 were Hispanic or Latino (4.2%). Patients who were Black were significantly younger, more were female, had later stage disease, were of lower socioeconomic status (SES), and were more likely to have unmutated immunoglobulin heavy chain gene (IGHV) and to have received cBTKi therapy than White patients (all p ≤ 0.002). SES was also significantly different by ethnicity. TTNT-D and OS were not different by race in either unadjusted or adjusted analyses (all p > 0.05). Conclusion: In unadjusted and adjusted analyses, TTNT-D and OS were not different by race. These data did not identify racial healthcare disparities in the era following the introduction of cBTKi therapy despite differences in baseline characteristics.

Chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL, hereafter CLL) is a cancer that is most commonly diagnosed among patients of White race in the USA. According to Surveillance, Epidemiology and End Results (SEER) data, the incidence rate of CLL in 2020 was 4.3 per 100,000 among individuals of White race and only 2.7 per 100,000 among those who are Black [1]. Although historically, Black patients with CLL have had significantly shorter overall survival (OS) when compared to White patients, the relative infrequency of diagnosis, compounded with the lower rates of enrollment to clinical trials, has limited the information available about patient outcomes for all other races in the current era of targeted therapies [2, 3].

BTK is a member of the TEC (tyrosine kinase expressed in hepatocellular carcinoma) family of nonreceptor tyrosine kinases and a key component of the B-cell receptor signaling complex. The B-cell receptor is required for normal and malignant B-cell maturation, proliferation, and survival [4, 5]. In 2014, the first covalent BTKi (cBTKi) was approved in the USA for the care of patients with CLL. Since then, there have been a total of three cBTKi agents approved by the US Food and Drug Administration (ibrutinib, zanubrutinib, and acalabrutinib). Pirtobrutinib, a noncovalent BTKi, has been approved for patients with CLL who have received at least two lines of therapy, including a cBTKi and a B-cell lymphoma 2 inhibitor (BCL2i) [6, 7]. From 2013 to 2016, the annual mortality rates for CLL decreased by 5.5%, generally corresponding to the introduction and adoption of these therapies [1]. In 2019, the BCL2i, venetoclax, was also approved for the care of patients with CLL, adding to the targeted therapies [8]. These agents have changed the landscape for patient care since the introduction of cBTKi therapy in 2014.

A study of outcomes in CLL patients by race conducted from 2004 to 2018 found that Black patients were younger, more likely to be uninsured, and to have more comorbidities [3]. While Black patients were more likely to receive immediate therapy than White patients, those who were Black had shorter OS than other races (median 7.0 years for Black patients vs. 9.0 for those who were White) [3]. Another study of patient outcomes early in the BTKi era (2015–2019) similarly found that there were differences in baseline characteristics such as younger age, worse Eastern Cooperative Oncology Group (ECOG) performance status, and more advanced stage at diagnosis among those who were Black; however, survival disparities were not observed by race, suggesting that these novel agents may have had a role in reducing healthcare disparities [9].

Given these intriguing data from the late 2010s suggesting the role of novel therapies in the reduction of survival disparities, this study was designed and conducted exclusively in the current time period to evaluate the hypotheses raised regarding race and survival outcomes in CLL after introduction and adoption of targeted therapy. Specifically, this study evaluated patient demographic and clinical characteristics, treatment patterns, and outcomes by both race and ethnicity, with a focus on the utilization of cBTKi therapies since their regulatory approval.

This study used the CLL Analytic Cohort from the nationwide Flatiron Health electronic health record-derived de-identified longitudinal database, comprising de-identified patient-level structured and unstructured data, curated via technology-enabled abstraction [10, 11]. De-identified data are not considered human subjects in accordance with US 45 CFR 46 [12]. Flatiron Health de-identified data originate from approximately 280 cancer clinics (∼800 sites of care) in the USA. Patients in the CLL Analytic Cohort are those who have CLL as identified by ICD-9: 204.1x or ICD-10: C91.1x or C83.0x codes, at least two documented clinical visits on different days occurring on or after January 1, 2011, and evidence of receiving antineoplastic therapy for CLL. Patients were included in the CLL Analytic Cohort if they received the initial CLL diagnosis on or after November 13, 2013 (the first approval date for cBTKi therapy) and had at least 121 days of follow-up in the database to allow for the observation of study outcomes.

Within the Flatiron Health database, race variables are collected as White, Black, Asian, other, or missing. Race in this study was evaluated for White, Black, and all non-White patients (including Black, Asian, and other race categories). Ethnicity is recorded as Hispanic or Latino, not Hispanic or Latino, or missing. Patients with missing race or ethnicity were excluded from these analyses and no imputation was made as to missing variables. However, those with missing race or ethnicity variables were retained in the study cohort and reported descriptively. Duration of cBTKi therapy was recorded in two ways. First, the duration of the initial cBTKi exposure was defined from the first use of cBTKi to the last use of cBTKi within a single line of therapy. Rechallenge of cBTKi in later lines of therapy was not included in this calculation. Second, the cumulative duration of cBTKi exposure was defined from the first use of cBTKi to the last use of cBTKi observed, excluding gap periods of 60 days or longer, to estimate the lifetime cumulative cBTKi exposure. Duration of therapy was estimated using Kaplan-Meier method; patients who were still receiving cBTKi therapy at the end of the database were censored at the patient’s last observation in the data. Lines of therapy reported in the Flatiron Health database were oncologist-defined and rule-based.

Outcomes evaluated included use of cBTKi therapy, time to next treatment or death (TTNT-D), and OS. Patients were considered to have received cBTKi therapy if they received ibrutinib, zanubrutinib, or acalabrutinib at any time on or after diagnosis; utilization was summarized descriptively by race and ethnicity. TTNT-D was defined as the time from the start of the line of therapy until the start of the immediate next line of therapy or death, whichever occurred first. If the time gap between the last follow-up and death date was less than or equal to 90 days, then that patient was considered to have an event for TTNT-D at the death date. Otherwise, patients were censored at the last follow-up date. OS was evaluated from the start of first-line therapy as well as from initial diagnosis, until time of death. Unadjusted time-to-event analyses were conducted using the unadjusted Kaplan-Meier method. Analyses were also conducted using a Cox proportional hazards regression model, adjusting for baseline covariates: year of CLL diagnosis, age at diagnosis, sex, race, ethnicity, site of care (community vs. academic practice), socioeconomic status, practice size (total number of patients with CLL treated at the site of care during the year of the patient’s index diagnosis), insurance type, and del(17p) status. Two interactions were also explored in the Cox proportional hazards regression model for OS. The first interaction was between ethnicity and site of care and the second interaction was between race and site of care while retaining all other covariates in the model. Patients without events were censored at the last activity date in the database. All statistical tests were conducted at a 5% significance level. All analyses were conducted using SAS version 9.4.

The statistical analysis plan included both unadjusted and adjusted analyses for the three primary study outcomes: use of cBTKi therapy, TTNT-D, and OS. Baseline characteristics were presented descriptively and compared using χ2 or t tests for categorical or continuous variables. Comparisons were conducted for White versus Black race, White versus all non-White races (including Black, Asian, and other race categories), and Hispanic or Latino versus not Hispanic or Latino. As mentioned previously, patients with missing race were excluded from the comparative analyses for race, and patients with missing ethnicity data were excluded for the comparisons by Hispanic or Latino ethnicity.

A total of 6,372 patients with CLL met eligibility criteria and were included in this study (shown in Fig. 1). White patients represented 71.8% of the cohort (n = 4,572), Black represented 8.8% (n = 558) of the cohort, and 18.5% were non-White (n = 1,180). Race was missing for 620 (9.7%). A small proportion of patients were Hispanic or Latino (n = 270; 4.2%), while most patients were not Hispanic or Latino (n = 4,897; 76.9%). Ethnicity was missing for 1,205 (18.9%). A summary of patient characteristics by race and ethnicity, including all covariates used in the comparative analyses, is included in Tables 1 and 2. Patients who were Black were significantly younger, more likely to be female, of lower socioeconomic status, and more likely to have unmutated IGHV gene and del(11q) than White patients. Patients who were Black also had a shorter time from diagnosis to initiation of first-line therapy. White patients were more likely to harbor the prognostically more favorable del(13q) molecular subtype. Patients who were Hispanic or Latino were significantly younger and had lower SES, but no significant differences were observed for sex or molecular subtypes, including IGHV mutation status. Both patients who were Black and those who were Hispanic or Latino were significantly more likely to be treated in centers with a lower total patient volume and less likely to receive care in an academic practice setting.

Fig. 1.

Cohort selection.

Fig. 1.

Cohort selection.

Close modal
Table 1.

Characteristics of study cohort by race

VariableAll patients (N = 6,372)White (N = 4,572)Black (N = 558)Non-Whitea (N = 1,180)Missing race (N = 620)p value, White versus Blackbp value, White versus non-Whiteb
Age at diagnosis 
 Mean (SD) 67.7 (10.2) 68.1 (9.9) 64.6 (11.0) 65.9 (10.6) 68.4 (10.7) <0.0001 <0.0001 
 Median (IQR) 69.0 (61.0, 77.0) 69.0 (61.0, 77.0) 66.0 (57.0, 73.0) 67.0 (59.0, 74.0) 71.0 (61.0, 77.0)   
Year of diagnosis, n (%) 
 2013–2014 1,056 (16.6) 819 (17.9) 81 (14.5) 166 (14.1) 71 (11.5) 0.05 <0.0001 
 2015 980 (15.4) 739 (16.2) 76 (13.6) 161 (13.6) 80 (12.9) 
 2016 991 (15.6) 734 (16.1) 83 (14.9) 172 (14.6) 85 (13.7) 
 2017 896 (14.1) 633 (13.9) 85 (15.2) 183 (15.5) 80 (12.9) 
 2018 811 (12.7) 564 (12.3) 75 (13.4) 152 (12.9) 95 (15.3) 
 2019 694 (10.9) 486 (10.6) 60 (10.8) 128 (10.9) 80 (12.9) 
 2020 535 (8.4) 347 (7.6) 54 (9.7) 127 (10.8) 61 (9.8) 
 2021 348 (5.5) 212 (4.6) 35 (6.3) 77 (6.5) 59 (9.5) 
 2022 61 (1.0) 38 (0.8) 9 (1.6) 14 (1.2) 9 (1.5) 
Sex, n (%) 
 Female 2,302 (36.1) 1,621 (35.5) 236 (42.3) 458 (38.8) 223 (36.0) 0.002 0.03 
 Male 4,070 (63.9) 2,951 (64.6) 322 (57.7) 722 (61.2) 397 (64.0) 
Geographic region, n (%) 
 Midwest 883 (13.9) 705 (15.4) 43 (7.7) 136 (11.5) 42 (6.8) <0.0001 <0.0001 
 Northeast 984 (15.4) 770 (16.8) 57 (10.2) 124 (10.5) 90 (14.5) 
 South or Puerto Rico 2,604 (40.9) 1,751 (38.3) 376 (67.4) 583 (49.4) 270 (43.6) 
 West 975 (15.3) 561 (12.3) 20 (3.6) 247 (20.9) 167 (26.9) 
 Missing 926 (14.5) 785 (17.2) 62 (11.1) 90 (7.6) 51 (8.2)   
Missing history of prior treatment for CLL, n (%)c 
 No 5,988 (94.0) 4,334 (94.8) 516 (92.5) 1,092 (92.5) 562 (90.7) 0.02 0.003 
 Yes 384 (6.0) 238 (5.2) 42 (7.5) 88 (7.5) 58 (9.4) 
Yost Index, n(%)d 
 1 – lowest SES 806 (12.7) 454 (9.9) 173 (31.0) 284 (24.1) 68 (11.0) <0.0001 <0.0001 
 2 978 (15.4) 676 (14.8) 114 (20.4) 212 (18.0) 90 (14.5) 
 3 1,117 (17.5) 803 (17.6) 71 (12.7) 178 (15.1) 136 (21.9) 
 4 1,156 (18.1) 844 (18.5) 69 (12.4) 190 (16.1) 122 (19.7) 
 5 – highest SES 1,038 (16.3) 779 (17.0) 40 (7.2) 138 (11.7) 121 (19.5) 
 Missing 1,277 (20.0) 1,016 (22.2) 91 (16.3) 178 (15.1) 83 (13.4)   
Year of first observed cBTKi, n (%) 
 2014 26 (0.4) 21 (0.5) 0.0 (0.0) 3 (0.3) 2 (0.3) 0.54 0.78 
 2015 99 (1.6) 72 (1.6) 8 (1.4) 18 (1.5) 9 (1.5) 
 2016 267 (4.2) 200 (4.4) 29 (5.2) 53 (4.5) 14 (2.3) 
 2017 364 (5.7) 256 (5.6) 42 (7.5) 84 (7.1) 24 (3.9) 
 2018 475 (7.5) 336 (7.4) 53 (9.5) 95 (8.1) 44 (7.1) 
 2019 692 (10.9) 494 (10.8) 81 (14.5) 137 (11.6) 61 (9.8) 
 2020 745 (11.7) 514 (11.2) 79 (14.2) 155 (13.1) 76 (12.3) 
 2021 642 (10.1) 457 (10.0) 60 (10.8) 123 (10.4) 62 (10.0) 
 2022 281 (4.4) 203 (4.4) 24 (4.3) 49 (4.2) 29 (4.7) 
 No cBTKi observed in the database 2,781 (43.6) 2,019 (44.2) 182 (32.6) 463 (39.2) 299 (48.2)   
BCL2i therapy, n (%) 
 No 5,394 (84.7) 3,865 (84.5) 475 (85.1) 992 (84.1) 537 (86.6) 0.72 0.69 
 Yes 978 (15.3) 707 (15.5) 83 (14.9) 188 (15.9) 83 (13.4) 
PI3Ki therapy, n (%) 
 No 6,320 (99.2) 4,536 (99.2) 549 (98.4) 1,165 (98.7) 619 (99.8) 0.06 0.11 
 Yes 52 (0.8) 36 (0.8) 9 (1.6) 15 (1.3) 1 (0.2) 
ECOG PS at diagnosis, n (%) 
 0 2,748 (43.1) 1,983 (43.4) 267 (47.9) 517 (43.8) 248 (40.0) 0.04 0.41 
 1 1,988 (31.2) 1,423 (31.1) 172 (30.8) 372 (31.5) 193 (31.1) 
 2 459 (7.2) 327 (7.2) 32 (5.7) 76 (6.4) 56 (9.0) 
 3 98 (1.5) 71 (1.6) 13 (2.3) 20 (1.7) 7 (1.1) 
 4 10 (0.2) 5 (0.1) 3 (0.5) 4 (0.3) 1 (0.2) 
 Missing 1,069 (16.8) 763 (16.7) 71 (12.7) 191 (16.2) 115 (18.6)   
ECOG PS at the start of first-line therapy, n (%) 
 0 2,224 (34.9) 1,630 (35.7) 199 (35.7) 408 (34.6) 186 (30.0) 0.002 0.02 
 1 1,634 (25.6) 1,153 (25.2) 147 (26.3) 325 (27.5) 156 (25.2) 
 2 340 (5.3) 232 (5.1) 23 (4.1) 64 (5.4) 44 (7.1) 
 3 70 (1.1) 55 (1.2) 5 (0.9) 11 (0.9) 4 (0.7) 
 4 8 (0.1) 3 (0.1) 4 (0.7) 5 (0.4) 0 (0.0) 
 Missing 2,096 (32.9) 1,499 (32.8) 180 (32.3) 367 (31.1) 230 (37.1)   
Disease subtype, n (%) 
 CLL 4,790 (75.2) 3,446 (75.4) 403 (72.2) 884 (74.9) 460 (74.2) 0.20 0.95 
 CLL/SLL 937 (14.7) 663 (14.5) 96 (17.2) 174 (14.8) 100 (16.1) 
 SLL 645 (10.1) 463 (10.1) 59 (10.6) 122 (10.3) 60 (9.7) 
Rai stage at diagnosis, n (%) 
 0 1,128 (17.7) 859 (18.8) 59 (10.6) 168 (14.2) 101 (16.3) <0.0001 0.06 
 I 869 (13.6) 638 (14.0) 89 (16.0) 160 (13.6) 71 (11.5) 
 II 484 (7.6) 352 (7.7) 33 (5.9) 87 (7.4) 45 (7.3) 
 III 465 (7.3) 321 (7.0) 53 (9.5) 93 (7.9) 51 (8.2) 
 IV 758 (11.9) 547 (12.0) 64 (11.5) 139 (11.8) 72 (11.6) 
 Missing 2,668 (41.9) 1,855 (40.6) 260 (46.6) 533 (45.2) 280 (45.2)   
Deletion11q, n (%) 
 Absent 3,671 (57.6) 2,676 (58.5) 285 (51.1) 647 (54.8) 348 (56.1) <0.0001 0.0001 
 Present 1,034 (16.2) 703 (15.4) 143 (25.6) 237 (20.1) 94 (15.2) 
 Missing 1,667 (26.2) 1,193 (26.1) 130 (23.3) 296 (25.1) 178 (28.7)   
Deletion13q, n (%) 
 Absent 2,422 (38.0) 1,677 (36.7) 274 (49.1) 520 (44.1) 225 (36.3) <0.0001 <0.0001 
 Present 2,402 (37.7) 1,805 (39.5) 146 (26.2) 371 (31.4) 226 (36.5) 
 Missing 1,548 (24.3) 1,090 (23.8) 138 (24.7) 289 (24.5) 169 (27.3)   
Deletion17p, n (%) 
 Absent 4,021 (63.1) 2880 (63.0) 367 (65.8) 765 (64.8) 376 (60.7) 0.18 0.48 
 Present 718 (11.3) 520 (11.4) 54 (9.7) 128 (10.9) 70 (11.3) 
 Missing 1,633 (25.6) 1,172 (25.6) 137 (24.6) 287 (24.3) 174 (28.1)   
IGHV, n (%) 
 Mutated 964 (15.1) 724 (15.8) 45 (8.1) 141 (12.0) 99 (16.0) <0.0001 0.002 
 Unmutated 1,685 (26.4) 1,214 (26.6) 200 (35.8) 334 (28.3) 137 (22.1) 
 Missing 3,723 (58.4) 2,634 (57.6) 313 (56.1) 705 (59.8) 384 (61.9)   
Payer type, n (%)e 
 Private 2,573 (40.4) 1,844 (40.3) 259 (46.4) 495 (42.0) 234 (37.7) 0.001 <0.0001 
 Public 2,075 (32.6) 1,552 (34.0) 158 (28.3) 335 (28.4) 188 (30.3) 
 Other 674 (10.6) 429 (9.4) 70 (12.5) 169 (14.3) 76 (12.3) 
 Missing 1,050 (16.5) 747 (16.3) 71 (12.7) 181 (15.3) 122 (19.7)   
Site of care, n (%) 
 Academic 808 (12.7) 694 (15.2) 58 (10.4) 80 (6.8) 34 (5.5) 0.0007 <0.0001 
 Community 5,488 (86.1) 3,809 (83.3) 498 (89.3) 1,093 (92.6) 586 (94.5) 
 Both 76 (1.2) 69 (1.5) 2 (0.4) 7 (0.6)  
Practice size, total number of patients with CLL per year 
 Evaluable patientsf 5,381 3,916 477 989 476   
 Mean (SD) 139.5 (114.6) 149.2 (119.4) 135.5 (114.8) 117.4 (98.8) 106.0 (90.2) 0.02 <0.0001 
 Median (IQR) 121.0 (45.0, 170.0) 121.0 (52.0, 181.0) 124.0 (45.0, 169.0) 97.0 (41.0, 169.0) 97.0 (32.0, 162.0)   
Time from diagnosis to first-line treatment, months 
 Evaluable patientsg 5,791 4,201 500 1,059 531   
 Mean (SD) 17.6 (20.1) 18.5 (20.4) 14.4 (18.6) 14.9 (19.1) 16.3 (19.0) <0.0001 <0.0001 
 Median (IQR) 9.2 (1.7, 27.7) 10.5 (1.8, 29.4) 4.8 (1.4, 22.7) 5.0 (1.5, 23.5) 8.3 (1.6, 25.4)   
Duration of follow-up, months 
 Mean (SD) 51.8 (27.5) 53.6 (27.53) 51.4 (27.7) 48.9 (26.9) 44.7 (27.0) 0.08 <0.0001 
 Median (IQR) 50.9 (28.9, 73.8) 53.2 (30.9, 75.8) 48.9 (28.9, 72.8) 46.6 (27.0, 69.0) 40.2 (20.9, 65.6)   
Total lines of therapy received 
 Evaluable patientsf 5,791 4,201 500 1,059 531   
 Mean (SD) 1.6 (1.1) 1.6 (1.1) 1.7 (1.1) 1.6 (1.1) 1.5 (0.8) 0.72 0.58 
 Median (IQR) 1.0 (1.0, 2.0) 1.0 (1.0, 2.0) 1.0 (1.0, 2.0) 1.0 (1.0, 2.0) 1.0 (1.0, 2.0)   
VariableAll patients (N = 6,372)White (N = 4,572)Black (N = 558)Non-Whitea (N = 1,180)Missing race (N = 620)p value, White versus Blackbp value, White versus non-Whiteb
Age at diagnosis 
 Mean (SD) 67.7 (10.2) 68.1 (9.9) 64.6 (11.0) 65.9 (10.6) 68.4 (10.7) <0.0001 <0.0001 
 Median (IQR) 69.0 (61.0, 77.0) 69.0 (61.0, 77.0) 66.0 (57.0, 73.0) 67.0 (59.0, 74.0) 71.0 (61.0, 77.0)   
Year of diagnosis, n (%) 
 2013–2014 1,056 (16.6) 819 (17.9) 81 (14.5) 166 (14.1) 71 (11.5) 0.05 <0.0001 
 2015 980 (15.4) 739 (16.2) 76 (13.6) 161 (13.6) 80 (12.9) 
 2016 991 (15.6) 734 (16.1) 83 (14.9) 172 (14.6) 85 (13.7) 
 2017 896 (14.1) 633 (13.9) 85 (15.2) 183 (15.5) 80 (12.9) 
 2018 811 (12.7) 564 (12.3) 75 (13.4) 152 (12.9) 95 (15.3) 
 2019 694 (10.9) 486 (10.6) 60 (10.8) 128 (10.9) 80 (12.9) 
 2020 535 (8.4) 347 (7.6) 54 (9.7) 127 (10.8) 61 (9.8) 
 2021 348 (5.5) 212 (4.6) 35 (6.3) 77 (6.5) 59 (9.5) 
 2022 61 (1.0) 38 (0.8) 9 (1.6) 14 (1.2) 9 (1.5) 
Sex, n (%) 
 Female 2,302 (36.1) 1,621 (35.5) 236 (42.3) 458 (38.8) 223 (36.0) 0.002 0.03 
 Male 4,070 (63.9) 2,951 (64.6) 322 (57.7) 722 (61.2) 397 (64.0) 
Geographic region, n (%) 
 Midwest 883 (13.9) 705 (15.4) 43 (7.7) 136 (11.5) 42 (6.8) <0.0001 <0.0001 
 Northeast 984 (15.4) 770 (16.8) 57 (10.2) 124 (10.5) 90 (14.5) 
 South or Puerto Rico 2,604 (40.9) 1,751 (38.3) 376 (67.4) 583 (49.4) 270 (43.6) 
 West 975 (15.3) 561 (12.3) 20 (3.6) 247 (20.9) 167 (26.9) 
 Missing 926 (14.5) 785 (17.2) 62 (11.1) 90 (7.6) 51 (8.2)   
Missing history of prior treatment for CLL, n (%)c 
 No 5,988 (94.0) 4,334 (94.8) 516 (92.5) 1,092 (92.5) 562 (90.7) 0.02 0.003 
 Yes 384 (6.0) 238 (5.2) 42 (7.5) 88 (7.5) 58 (9.4) 
Yost Index, n(%)d 
 1 – lowest SES 806 (12.7) 454 (9.9) 173 (31.0) 284 (24.1) 68 (11.0) <0.0001 <0.0001 
 2 978 (15.4) 676 (14.8) 114 (20.4) 212 (18.0) 90 (14.5) 
 3 1,117 (17.5) 803 (17.6) 71 (12.7) 178 (15.1) 136 (21.9) 
 4 1,156 (18.1) 844 (18.5) 69 (12.4) 190 (16.1) 122 (19.7) 
 5 – highest SES 1,038 (16.3) 779 (17.0) 40 (7.2) 138 (11.7) 121 (19.5) 
 Missing 1,277 (20.0) 1,016 (22.2) 91 (16.3) 178 (15.1) 83 (13.4)   
Year of first observed cBTKi, n (%) 
 2014 26 (0.4) 21 (0.5) 0.0 (0.0) 3 (0.3) 2 (0.3) 0.54 0.78 
 2015 99 (1.6) 72 (1.6) 8 (1.4) 18 (1.5) 9 (1.5) 
 2016 267 (4.2) 200 (4.4) 29 (5.2) 53 (4.5) 14 (2.3) 
 2017 364 (5.7) 256 (5.6) 42 (7.5) 84 (7.1) 24 (3.9) 
 2018 475 (7.5) 336 (7.4) 53 (9.5) 95 (8.1) 44 (7.1) 
 2019 692 (10.9) 494 (10.8) 81 (14.5) 137 (11.6) 61 (9.8) 
 2020 745 (11.7) 514 (11.2) 79 (14.2) 155 (13.1) 76 (12.3) 
 2021 642 (10.1) 457 (10.0) 60 (10.8) 123 (10.4) 62 (10.0) 
 2022 281 (4.4) 203 (4.4) 24 (4.3) 49 (4.2) 29 (4.7) 
 No cBTKi observed in the database 2,781 (43.6) 2,019 (44.2) 182 (32.6) 463 (39.2) 299 (48.2)   
BCL2i therapy, n (%) 
 No 5,394 (84.7) 3,865 (84.5) 475 (85.1) 992 (84.1) 537 (86.6) 0.72 0.69 
 Yes 978 (15.3) 707 (15.5) 83 (14.9) 188 (15.9) 83 (13.4) 
PI3Ki therapy, n (%) 
 No 6,320 (99.2) 4,536 (99.2) 549 (98.4) 1,165 (98.7) 619 (99.8) 0.06 0.11 
 Yes 52 (0.8) 36 (0.8) 9 (1.6) 15 (1.3) 1 (0.2) 
ECOG PS at diagnosis, n (%) 
 0 2,748 (43.1) 1,983 (43.4) 267 (47.9) 517 (43.8) 248 (40.0) 0.04 0.41 
 1 1,988 (31.2) 1,423 (31.1) 172 (30.8) 372 (31.5) 193 (31.1) 
 2 459 (7.2) 327 (7.2) 32 (5.7) 76 (6.4) 56 (9.0) 
 3 98 (1.5) 71 (1.6) 13 (2.3) 20 (1.7) 7 (1.1) 
 4 10 (0.2) 5 (0.1) 3 (0.5) 4 (0.3) 1 (0.2) 
 Missing 1,069 (16.8) 763 (16.7) 71 (12.7) 191 (16.2) 115 (18.6)   
ECOG PS at the start of first-line therapy, n (%) 
 0 2,224 (34.9) 1,630 (35.7) 199 (35.7) 408 (34.6) 186 (30.0) 0.002 0.02 
 1 1,634 (25.6) 1,153 (25.2) 147 (26.3) 325 (27.5) 156 (25.2) 
 2 340 (5.3) 232 (5.1) 23 (4.1) 64 (5.4) 44 (7.1) 
 3 70 (1.1) 55 (1.2) 5 (0.9) 11 (0.9) 4 (0.7) 
 4 8 (0.1) 3 (0.1) 4 (0.7) 5 (0.4) 0 (0.0) 
 Missing 2,096 (32.9) 1,499 (32.8) 180 (32.3) 367 (31.1) 230 (37.1)   
Disease subtype, n (%) 
 CLL 4,790 (75.2) 3,446 (75.4) 403 (72.2) 884 (74.9) 460 (74.2) 0.20 0.95 
 CLL/SLL 937 (14.7) 663 (14.5) 96 (17.2) 174 (14.8) 100 (16.1) 
 SLL 645 (10.1) 463 (10.1) 59 (10.6) 122 (10.3) 60 (9.7) 
Rai stage at diagnosis, n (%) 
 0 1,128 (17.7) 859 (18.8) 59 (10.6) 168 (14.2) 101 (16.3) <0.0001 0.06 
 I 869 (13.6) 638 (14.0) 89 (16.0) 160 (13.6) 71 (11.5) 
 II 484 (7.6) 352 (7.7) 33 (5.9) 87 (7.4) 45 (7.3) 
 III 465 (7.3) 321 (7.0) 53 (9.5) 93 (7.9) 51 (8.2) 
 IV 758 (11.9) 547 (12.0) 64 (11.5) 139 (11.8) 72 (11.6) 
 Missing 2,668 (41.9) 1,855 (40.6) 260 (46.6) 533 (45.2) 280 (45.2)   
Deletion11q, n (%) 
 Absent 3,671 (57.6) 2,676 (58.5) 285 (51.1) 647 (54.8) 348 (56.1) <0.0001 0.0001 
 Present 1,034 (16.2) 703 (15.4) 143 (25.6) 237 (20.1) 94 (15.2) 
 Missing 1,667 (26.2) 1,193 (26.1) 130 (23.3) 296 (25.1) 178 (28.7)   
Deletion13q, n (%) 
 Absent 2,422 (38.0) 1,677 (36.7) 274 (49.1) 520 (44.1) 225 (36.3) <0.0001 <0.0001 
 Present 2,402 (37.7) 1,805 (39.5) 146 (26.2) 371 (31.4) 226 (36.5) 
 Missing 1,548 (24.3) 1,090 (23.8) 138 (24.7) 289 (24.5) 169 (27.3)   
Deletion17p, n (%) 
 Absent 4,021 (63.1) 2880 (63.0) 367 (65.8) 765 (64.8) 376 (60.7) 0.18 0.48 
 Present 718 (11.3) 520 (11.4) 54 (9.7) 128 (10.9) 70 (11.3) 
 Missing 1,633 (25.6) 1,172 (25.6) 137 (24.6) 287 (24.3) 174 (28.1)   
IGHV, n (%) 
 Mutated 964 (15.1) 724 (15.8) 45 (8.1) 141 (12.0) 99 (16.0) <0.0001 0.002 
 Unmutated 1,685 (26.4) 1,214 (26.6) 200 (35.8) 334 (28.3) 137 (22.1) 
 Missing 3,723 (58.4) 2,634 (57.6) 313 (56.1) 705 (59.8) 384 (61.9)   
Payer type, n (%)e 
 Private 2,573 (40.4) 1,844 (40.3) 259 (46.4) 495 (42.0) 234 (37.7) 0.001 <0.0001 
 Public 2,075 (32.6) 1,552 (34.0) 158 (28.3) 335 (28.4) 188 (30.3) 
 Other 674 (10.6) 429 (9.4) 70 (12.5) 169 (14.3) 76 (12.3) 
 Missing 1,050 (16.5) 747 (16.3) 71 (12.7) 181 (15.3) 122 (19.7)   
Site of care, n (%) 
 Academic 808 (12.7) 694 (15.2) 58 (10.4) 80 (6.8) 34 (5.5) 0.0007 <0.0001 
 Community 5,488 (86.1) 3,809 (83.3) 498 (89.3) 1,093 (92.6) 586 (94.5) 
 Both 76 (1.2) 69 (1.5) 2 (0.4) 7 (0.6)  
Practice size, total number of patients with CLL per year 
 Evaluable patientsf 5,381 3,916 477 989 476   
 Mean (SD) 139.5 (114.6) 149.2 (119.4) 135.5 (114.8) 117.4 (98.8) 106.0 (90.2) 0.02 <0.0001 
 Median (IQR) 121.0 (45.0, 170.0) 121.0 (52.0, 181.0) 124.0 (45.0, 169.0) 97.0 (41.0, 169.0) 97.0 (32.0, 162.0)   
Time from diagnosis to first-line treatment, months 
 Evaluable patientsg 5,791 4,201 500 1,059 531   
 Mean (SD) 17.6 (20.1) 18.5 (20.4) 14.4 (18.6) 14.9 (19.1) 16.3 (19.0) <0.0001 <0.0001 
 Median (IQR) 9.2 (1.7, 27.7) 10.5 (1.8, 29.4) 4.8 (1.4, 22.7) 5.0 (1.5, 23.5) 8.3 (1.6, 25.4)   
Duration of follow-up, months 
 Mean (SD) 51.8 (27.5) 53.6 (27.53) 51.4 (27.7) 48.9 (26.9) 44.7 (27.0) 0.08 <0.0001 
 Median (IQR) 50.9 (28.9, 73.8) 53.2 (30.9, 75.8) 48.9 (28.9, 72.8) 46.6 (27.0, 69.0) 40.2 (20.9, 65.6)   
Total lines of therapy received 
 Evaluable patientsf 5,791 4,201 500 1,059 531   
 Mean (SD) 1.6 (1.1) 1.6 (1.1) 1.7 (1.1) 1.6 (1.1) 1.5 (0.8) 0.72 0.58 
 Median (IQR) 1.0 (1.0, 2.0) 1.0 (1.0, 2.0) 1.0 (1.0, 2.0) 1.0 (1.0, 2.0) 1.0 (1.0, 2.0)   

SD, standard deviation; IQR, interquartile range; ECOG PS, Eastern Cooperative Oncology Group performance status; SES, socioeconomic status; IGHV, immunoglobulin heavy-chain variable region gene.

aNon-White includes Black, Asian, and other nonspecified race categories, excluding those with missing race.

bp values calculated by χ2 test for categorical variables, excluding missing categories, and by t test for continuous variables.

cHistory of treatment for CLL that is not documented in the database, potentially due to care prior to referral to a center where Flatiron Health collects data; therefore, prior lines of therapy and receipt of prior treatments are unknown.

dThe Yost Index23 is a composite measure of socioeconomic status, incorporating regional variables such as median household income, median home value, median gross rent, percentage of individuals living below 150% of the poverty line, percentage of individuals considered working class, percentage of individuals who are unemployed, and education index and is not a directly obtained variable from the patient or the provider.

ePrivate includes patients with any combination of plans that include at least one private payer (with or without a public payer component).

fSite of care details were not available for all patients in the study cohort.

gLimited to patients where the date of first-line therapy was known, excluding those missing history of prior treatment.

Table 2.

Characteristics of study cohort by ethnicity

VariableHispanic or Latino (N = 270)Not Hispanic or Latino (N = 4,897)Missing ethnicity (N = 1,205)p value, Hispanic or Latino versus not Hispanic or Latinoa
Age at diagnosis 
 Mean (SD) 66.1 (10.7) 67.7 (10.1) 68.1 (10.5) 0.01 
 Median (IQR) 67.5 (59.0, 75.0) 69.0 (61.0, 76.0) 70.0 (61.0, 77.0)  
Year of diagnosis, n (%) 
 2013–2014 41 (15.2) 880 (18.0) 135 (11.2) 0.02 
 2015 35 (13.0) 804 (16.4) 141 (11.7) 
 2016 38 (14.1) 802 (16.4) 151 (12.5) 
 2017 42 (15.6) 693 (14.2) 161 (13.4) 
 2018 28 (10.4) 610 (12.5) 173 (14.4) 
 2019 35 (13.0) 484 (9.9) 175 (14.5) 
 2020 28 (10.4) 361 (7.4) 146 (12.1) 
 2021 22 (8.2) 218 (4.5) 108 (9.0) 
 2022 1 (0.4) 45 (0.9) 15 (1.2) 
Sex, n (%) 
 Female 106 (39.3) 1,747 (35.7) 449 (37.3) 0.23 
 Male 164 (60.7) 3,150 (64.3) 756 (62.7) 
Geographic region, n (%) 
 Midwest 9 (3.3) 725 (14.8) 149 (12.4) <0.0001 
 Northeast 28 (10.4) 800 (16.3) 156 (13.0) 
 South of Puerto Rico 137 (50.7) 1,892 (38.6) 575 (47.7) 
 West 66 (24.4) 622 (12.7) 287 (23.8) 
 Missing 30 (11.1) 858 (17.5) 38 (3.2)  
Missing history of prior treatment for CLL, n (%)b 
 No 253 (93.7) 4,627 (94.5) 1,108 (92.0) 0.58 
 Yes 17 (6.3) 270 (5.5) 97 (8.1) 
Yost Index, n(%)c 
 1 – lowest SES 82 (30.4) 558 (11.4) 166 (13.8) <0.0001 
 2 40 (14.8) 732 (15.0) 206 (17.1) 
 3 44 (16.3) 834 (17.0) 239 (19.8) 
 4 27 (10.0) 891 (18.2) 238 (19.8) 
 5 – highest SES 16 (5.9) 801 (16.4) 221 (18.3) 
 Missing 61 (22.6) 1,081 (22.1) 135 (11.2  
BCL2i therapy, n (%) 
 No 226 (83.7) 4,169 (85.1) 999 (82.9) 0.52 
 Yes 4 (16.3) 728 (14.9) 206 (17.1) 
PI3Ki therapy, n (%) 
 No 265 (98.1) 4,855 (99.1) 1,200 (99.6) 0.10 
 Yes 5 (1.9) 42 (0.9) 5 (0.4) 
Year of first observed cBTKi, n (%) 
 2014 1 (0.4) 21 (0.4) 4 (0.3) 1.00 
 2015 5 (1.9) 81 (1.7) 13 (1.1) 
 2016 14 (5.2) 224 (4.6) 29 (2.4) 
 2017 15 (5.6) 292 (6.0) 57 (4.7) 
 2018 18 (6.7) 391 (8.0) 66 (5.5) 
 2019 27 (10.0) 531 (10.8) 134 (11.1) 
 2020 29 (10.7) 566 (11.6) 150 (12.5) 
 2021 25 (9.3) 482 (9.8) 135 (11.2) 
 2022 10 (3.7) 212 (4.3) 59 (4.9) 
 No cBTKi observed in the database 126 (46.7) 2,097 (42.8) 558 (46.3)  
ECOG PS at diagnosis, n (%) 
 0 116 (43.0) 2,075 (42.4) 557 (46.2) 0.90 
 1 80 (29.6) 1,528 (31.2) 380 (31.5) 
 2 19 (7.0) 343 (7.0) 97 (8.1) 
 3 3 (1.1) 82 (1.7) 13 (1.1) 
 4 0 (0.0) 7 (0.1) 3 (0.3) 
 Missing 52 (19.3) 862 (17.6) 155 (12.9)  
ECOG PS at the start of first-line therapy, n (%) 
 0 102 (37.8) 1,667 (34.0) 455 (37.8) 0.51 
 1 58 (21.5) 1,251 (25.6) 325 (27.0) 
 2 15 (5.6) 253 (5.2) 72 (6.0) 
 3 4 (1.5) 57 (1.2) 9 (0.8) 
 4 0 (0.0) 5 (0.1) 3 (0.3) 
 Missing 91 (33.7) 1,664 (34.0) 341 (28.3)  
Disease subtype, n (%) 
 CLL 199 (73.7) 3,676 (75.1) 915 (75.9) 0.67 
 CLL/SLL 39 (14.4) 724 (14.8) 174 (14.4) 
 SLL 32 (11.9) 497 (10.2) 116 (9.6) 
Rai stage at diagnosis, n (%) 
 0 33 (12.2) 891 (18.2) 204 (16.9) 0.09 
 I 24 (8.9) 701 (14.3) 144 (12.0) 
 II 23 (8.5) 370 (7.6) 91 (7.6) 
 III 23 (8.5) 366 (7.5) 76 (6.3) 
 IV 29 (10.7) 574 (11.7) 155 (12.9) 
 Missing 138 (51.1) 1,995 (40.7) 535 (44.4)  
Deletion11q, n (%) 
 Absent 150 (55.6) 2,832 (57.8) 689 (57.2) 0.88 
 Present 41 (15.2) 795 (16.2) 198 (16.4) 
 Missing 79 (29.3) 1,270 (25.9) 318 (26.4)  
Deletion13q, n (%) 
 Absent 102 (37.8) 1,876 (38.3) 444 (36.9) 0.62 
 Present 93 (34.4) 1,840 (37.6) 469 (38.9) 
 Missing 75 (27.8) 1,181 (24.1) 292 (24.2)  
Deletion17p, n (%) 
 Absent 163 (60.4) 3,091 (63.1) 767 (63.7) 0.99 
 Present 29 (10.7) 551 (11.3) 138 (11.5) 
 Missing 78 (28.9) 1,255 (25.6) 300 (24.9)  
IGHV, n (%) 
 Mutated 42 (15.6) 721 (14.7) 201 (16.7) 0.27 
 Unmutated 62 (23.0) 1,332 (27.2) 291 (24.2) 
 Missing 166 (61.5) 2,844 (58.1) 713 (59.2)  
Payer type, n (%)d 
 Private 112 (41.5) 2010 (41.1) 451 (37.4) <0.0001 
 Public 57 (21.1) 1,642 (33.5) 376 (31.2) 
 Other 44 (16.3) 494 (10.1) 136 (11.3) 
 Missing 57 (21.1) 751 (15.3) 242 (20.1)  
Site of care, n (%) 
 Academic 28 (10.4) 759 (15.5) 21 (1.7) 0.02 
 Community 241 (89.3) 4,064 (83.0) 1,183 (98.2) 
 Both 1 (0.4) 74 (1.5) 1 (0.1) 
Practice size, total number of patients with CLL per year 
 Mean (SD) 77.5 (89.6) 147.6 (119.0) 118.39 (90.8) <0.0001 
 Median (IQR) 41.0 (9.0, 99.0) 121.0 (52.0, 181.0) 99.0 (41.0, 162.0)  
 Evaluable patientse 222 4,197 962  
Time from diagnosis to first-line treatment, months 
 Mean (SD) 15.2 (20.0) 18.1 (20.4) 16.1 (19.0) 0.03 
 Median (IQR) 4.0 (1.5, 24.5) 10.1 (1.8, 28.4) 7.2 (1.6, 25.0)  
 Evaluable patientsf 240 4,487 1,064  
Duration of follow-up, months 
 Mean (SD) 46.8 (27.1) 53.7 (27.5) 45.3 (26.5) <0.0001 
 Median (IQR) 44.0 (23.6, 66.6) 53.7 (31.1, 76.0) 41.0 (23.5, 65.0)  
VariableHispanic or Latino (N = 270)Not Hispanic or Latino (N = 4,897)Missing ethnicity (N = 1,205)p value, Hispanic or Latino versus not Hispanic or Latinoa
Age at diagnosis 
 Mean (SD) 66.1 (10.7) 67.7 (10.1) 68.1 (10.5) 0.01 
 Median (IQR) 67.5 (59.0, 75.0) 69.0 (61.0, 76.0) 70.0 (61.0, 77.0)  
Year of diagnosis, n (%) 
 2013–2014 41 (15.2) 880 (18.0) 135 (11.2) 0.02 
 2015 35 (13.0) 804 (16.4) 141 (11.7) 
 2016 38 (14.1) 802 (16.4) 151 (12.5) 
 2017 42 (15.6) 693 (14.2) 161 (13.4) 
 2018 28 (10.4) 610 (12.5) 173 (14.4) 
 2019 35 (13.0) 484 (9.9) 175 (14.5) 
 2020 28 (10.4) 361 (7.4) 146 (12.1) 
 2021 22 (8.2) 218 (4.5) 108 (9.0) 
 2022 1 (0.4) 45 (0.9) 15 (1.2) 
Sex, n (%) 
 Female 106 (39.3) 1,747 (35.7) 449 (37.3) 0.23 
 Male 164 (60.7) 3,150 (64.3) 756 (62.7) 
Geographic region, n (%) 
 Midwest 9 (3.3) 725 (14.8) 149 (12.4) <0.0001 
 Northeast 28 (10.4) 800 (16.3) 156 (13.0) 
 South of Puerto Rico 137 (50.7) 1,892 (38.6) 575 (47.7) 
 West 66 (24.4) 622 (12.7) 287 (23.8) 
 Missing 30 (11.1) 858 (17.5) 38 (3.2)  
Missing history of prior treatment for CLL, n (%)b 
 No 253 (93.7) 4,627 (94.5) 1,108 (92.0) 0.58 
 Yes 17 (6.3) 270 (5.5) 97 (8.1) 
Yost Index, n(%)c 
 1 – lowest SES 82 (30.4) 558 (11.4) 166 (13.8) <0.0001 
 2 40 (14.8) 732 (15.0) 206 (17.1) 
 3 44 (16.3) 834 (17.0) 239 (19.8) 
 4 27 (10.0) 891 (18.2) 238 (19.8) 
 5 – highest SES 16 (5.9) 801 (16.4) 221 (18.3) 
 Missing 61 (22.6) 1,081 (22.1) 135 (11.2  
BCL2i therapy, n (%) 
 No 226 (83.7) 4,169 (85.1) 999 (82.9) 0.52 
 Yes 4 (16.3) 728 (14.9) 206 (17.1) 
PI3Ki therapy, n (%) 
 No 265 (98.1) 4,855 (99.1) 1,200 (99.6) 0.10 
 Yes 5 (1.9) 42 (0.9) 5 (0.4) 
Year of first observed cBTKi, n (%) 
 2014 1 (0.4) 21 (0.4) 4 (0.3) 1.00 
 2015 5 (1.9) 81 (1.7) 13 (1.1) 
 2016 14 (5.2) 224 (4.6) 29 (2.4) 
 2017 15 (5.6) 292 (6.0) 57 (4.7) 
 2018 18 (6.7) 391 (8.0) 66 (5.5) 
 2019 27 (10.0) 531 (10.8) 134 (11.1) 
 2020 29 (10.7) 566 (11.6) 150 (12.5) 
 2021 25 (9.3) 482 (9.8) 135 (11.2) 
 2022 10 (3.7) 212 (4.3) 59 (4.9) 
 No cBTKi observed in the database 126 (46.7) 2,097 (42.8) 558 (46.3)  
ECOG PS at diagnosis, n (%) 
 0 116 (43.0) 2,075 (42.4) 557 (46.2) 0.90 
 1 80 (29.6) 1,528 (31.2) 380 (31.5) 
 2 19 (7.0) 343 (7.0) 97 (8.1) 
 3 3 (1.1) 82 (1.7) 13 (1.1) 
 4 0 (0.0) 7 (0.1) 3 (0.3) 
 Missing 52 (19.3) 862 (17.6) 155 (12.9)  
ECOG PS at the start of first-line therapy, n (%) 
 0 102 (37.8) 1,667 (34.0) 455 (37.8) 0.51 
 1 58 (21.5) 1,251 (25.6) 325 (27.0) 
 2 15 (5.6) 253 (5.2) 72 (6.0) 
 3 4 (1.5) 57 (1.2) 9 (0.8) 
 4 0 (0.0) 5 (0.1) 3 (0.3) 
 Missing 91 (33.7) 1,664 (34.0) 341 (28.3)  
Disease subtype, n (%) 
 CLL 199 (73.7) 3,676 (75.1) 915 (75.9) 0.67 
 CLL/SLL 39 (14.4) 724 (14.8) 174 (14.4) 
 SLL 32 (11.9) 497 (10.2) 116 (9.6) 
Rai stage at diagnosis, n (%) 
 0 33 (12.2) 891 (18.2) 204 (16.9) 0.09 
 I 24 (8.9) 701 (14.3) 144 (12.0) 
 II 23 (8.5) 370 (7.6) 91 (7.6) 
 III 23 (8.5) 366 (7.5) 76 (6.3) 
 IV 29 (10.7) 574 (11.7) 155 (12.9) 
 Missing 138 (51.1) 1,995 (40.7) 535 (44.4)  
Deletion11q, n (%) 
 Absent 150 (55.6) 2,832 (57.8) 689 (57.2) 0.88 
 Present 41 (15.2) 795 (16.2) 198 (16.4) 
 Missing 79 (29.3) 1,270 (25.9) 318 (26.4)  
Deletion13q, n (%) 
 Absent 102 (37.8) 1,876 (38.3) 444 (36.9) 0.62 
 Present 93 (34.4) 1,840 (37.6) 469 (38.9) 
 Missing 75 (27.8) 1,181 (24.1) 292 (24.2)  
Deletion17p, n (%) 
 Absent 163 (60.4) 3,091 (63.1) 767 (63.7) 0.99 
 Present 29 (10.7) 551 (11.3) 138 (11.5) 
 Missing 78 (28.9) 1,255 (25.6) 300 (24.9)  
IGHV, n (%) 
 Mutated 42 (15.6) 721 (14.7) 201 (16.7) 0.27 
 Unmutated 62 (23.0) 1,332 (27.2) 291 (24.2) 
 Missing 166 (61.5) 2,844 (58.1) 713 (59.2)  
Payer type, n (%)d 
 Private 112 (41.5) 2010 (41.1) 451 (37.4) <0.0001 
 Public 57 (21.1) 1,642 (33.5) 376 (31.2) 
 Other 44 (16.3) 494 (10.1) 136 (11.3) 
 Missing 57 (21.1) 751 (15.3) 242 (20.1)  
Site of care, n (%) 
 Academic 28 (10.4) 759 (15.5) 21 (1.7) 0.02 
 Community 241 (89.3) 4,064 (83.0) 1,183 (98.2) 
 Both 1 (0.4) 74 (1.5) 1 (0.1) 
Practice size, total number of patients with CLL per year 
 Mean (SD) 77.5 (89.6) 147.6 (119.0) 118.39 (90.8) <0.0001 
 Median (IQR) 41.0 (9.0, 99.0) 121.0 (52.0, 181.0) 99.0 (41.0, 162.0)  
 Evaluable patientse 222 4,197 962  
Time from diagnosis to first-line treatment, months 
 Mean (SD) 15.2 (20.0) 18.1 (20.4) 16.1 (19.0) 0.03 
 Median (IQR) 4.0 (1.5, 24.5) 10.1 (1.8, 28.4) 7.2 (1.6, 25.0)  
 Evaluable patientsf 240 4,487 1,064  
Duration of follow-up, months 
 Mean (SD) 46.8 (27.1) 53.7 (27.5) 45.3 (26.5) <0.0001 
 Median (IQR) 44.0 (23.6, 66.6) 53.7 (31.1, 76.0) 41.0 (23.5, 65.0)  

SD, standard deviation; IQR, interquartile range; ECOG PS, Eastern Cooperative Oncology Group performance status; SES, socioeconomic status; IGHV, immunoglobulin heavy-chain variable region genes.

ap values were calculated by the χ2 test for categorical variables, excluding missing categories, and by the t test for continuous variables.

bHistory of treatment for CLL that is not documented in the database, potentially due to care prior to referral to a center where Flatiron Health collects data; therefore, prior lines of therapy and receipt of prior treatments are unknown.

cThe Yost SES Index23 is a composite measure of socioeconomic status, incorporating regional variables such as median household income, median home value, median gross rent, percentage of individuals living below 150% of the poverty line, percentage of individuals considered working class, percentage of individuals who are unemployed, and education index and is not a directly obtained variable from the patient or the provider.

dPrivate includes patients with any combination of plans that include at least one private payer (with or without a public payer component).

eSite of care details were not available for all patients in the study cohort.

fLimited to patients where the date of first-line therapy was known, excluding those missing history of prior treatment.

Patterns of cBTKi Treatment

The increasing utilization of cBTKi therapy by the year of initiation (Fig. 2) was not significantly different by the racial group (Table 2). As shown in Table 3, patterns of cBTKi utilization were significantly different by race: 67.4% of Black versus 55.8% of White patients received cBTKi therapy (p < 0.0001). Non-White patients also had a significantly higher use of cBTKi therapy than White patients (60.8 vs. 55.8%, p = 0.002). No significant differences were observed by ethnicity (53.3 Hispanic/Latino vs. 57.2% not Hispanic/Latino, p = 0.21). There was less time from diagnosis to cBTKi therapy for Black and non-White patients versus White as well as for Hispanic or Latino patients versus those who were not Hispanic or Latino. There were significantly higher rates of cBTKi use in the first-line setting for Black patients than for those who were White (49.2 vs. 40.3%, p = 0.0001), as well as for those who were non-White versus those who were White (44.5 vs. 40.3% p = 0.01). Both patients who were Black as well as those who were not Hispanic or Latino had a longer duration of cumulative cBTKi therapy and a longer duration of first-line cBTKi therapy than those who were White or who were Hispanic or Latino, respectively (all p < 0.05). There were no differences by race for the year of the first observed cBTKi, nor were there differences by race for BCL2i or PI3Ki utilization (Table 2).

Fig. 2.

Cumulative proportion by the year of initiation of cBTKi therapy (n = 3,370).

Fig. 2.

Cumulative proportion by the year of initiation of cBTKi therapy (n = 3,370).

Close modal
Table 3.

BTKi exposure by race and ethnicity

VariableAll patients (N = 6,372)White (N = 4,572)Black (N = 558)Non-Whitea (N = 1,180)p valuebHispanic or Latino (N = 270)Not Hispanic or Latino (N = 4,897)Hispanic Latino versus not Hispanic Latino, p valueb
cBTKi exposure = yes, n (%)b 3,591 (56.4) 2,553 (55.8) 376 (67.4) 717 (60.8) White versus Black, p < 0.0001 144 (53.3) 2,800 (57.2) p = 0.21 
White versus non-White, p = 0.002 
VariableAll patients (N = 6,372)White (N = 4,572)Black (N = 558)Non-Whitea (N = 1,180)p valuebHispanic or Latino (N = 270)Not Hispanic or Latino (N = 4,897)Hispanic Latino versus not Hispanic Latino, p valueb
cBTKi exposure = yes, n (%)b 3,591 (56.4) 2,553 (55.8) 376 (67.4) 717 (60.8) White versus Black, p < 0.0001 144 (53.3) 2,800 (57.2) p = 0.21 
White versus non-White, p = 0.002 
All patientsa (N = 5,791)Whitea (N = 4,201)Blacka (N = 500)Non-Whitea (N = 1,059)pvaluebHispanic or Latinoa (N = 240)Not Hispanic or Latinoa (N = 4,487)Hispanic Latino versus not Hispanic Latino, p valueb
Initial cBTKi received in first line, n (%) 2374 (41.0) 1,691 (40.3) 246 (49.2) 471 (44.5) White versus Black, p = 0.0001 86 (35.8) 1,846 (41.1) p = 0.10 
White versus non-White, p = 0.01 
Initial cBTKi received in second line, n (%) 640 (11.1) 476 (11.3) 62 (12.4) 115 (10.9) White versus Black, p = 0.48 28 (11.7) 518 (11.5) p = 0.95 
White versus non-White, p = 0.66 
Initial cBTKi received in third or later line, n (%) 191 (3.30) 141 (3.4) 25 (5.0) 47 (4.4) White versus Black, p = 0.06 10 (4.2) 158 (3.5) p = 0.60 
White versus non-White, p = 0.09 
Time from diagnosis to initial cBTKi, median months (IQR) 19.9 (3.8, 41.9) 21.8 (4.9, 44.4) 15.4 (2.2, 36.0) 16.0 (2.2, 36.0) White versus Black, p = 0.0005 12.7 (2.7, 38.2) 21.4 (4.2, 43.6) p = 0.05 
White versus non-White, p < 0.0001 
Duration of initial cBTKi exposure, median (95% CI), months 15.7 (14.8, 16.5) 15.6 (14.5, 16.7) 21.8 (17.9, 23.5) 16.9 (15.5, 19.1) White versus Black, p = 0.03 13.3 (10.1, 16.1) 16.7 (15.7, 17.9) p<0.0001 
White versus non-White, p = 0.76 
Cumulative duration of cBTKi exposure, median (95% CI), months 35.1 (32.5, 37.0) 35.3 (31.8, 37.6) 41.7 (34.2, 51.0) 35.6 (32.1, 41.9) White versus Black, p = 0.01 25.1 (17.8, 46.1) 35.9 (33.6, 38.1) p = 0.03 
White versus non-White, p = 0.21 
All patientsa (N = 5,791)Whitea (N = 4,201)Blacka (N = 500)Non-Whitea (N = 1,059)pvaluebHispanic or Latinoa (N = 240)Not Hispanic or Latinoa (N = 4,487)Hispanic Latino versus not Hispanic Latino, p valueb
Initial cBTKi received in first line, n (%) 2374 (41.0) 1,691 (40.3) 246 (49.2) 471 (44.5) White versus Black, p = 0.0001 86 (35.8) 1,846 (41.1) p = 0.10 
White versus non-White, p = 0.01 
Initial cBTKi received in second line, n (%) 640 (11.1) 476 (11.3) 62 (12.4) 115 (10.9) White versus Black, p = 0.48 28 (11.7) 518 (11.5) p = 0.95 
White versus non-White, p = 0.66 
Initial cBTKi received in third or later line, n (%) 191 (3.30) 141 (3.4) 25 (5.0) 47 (4.4) White versus Black, p = 0.06 10 (4.2) 158 (3.5) p = 0.60 
White versus non-White, p = 0.09 
Time from diagnosis to initial cBTKi, median months (IQR) 19.9 (3.8, 41.9) 21.8 (4.9, 44.4) 15.4 (2.2, 36.0) 16.0 (2.2, 36.0) White versus Black, p = 0.0005 12.7 (2.7, 38.2) 21.4 (4.2, 43.6) p = 0.05 
White versus non-White, p < 0.0001 
Duration of initial cBTKi exposure, median (95% CI), months 15.7 (14.8, 16.5) 15.6 (14.5, 16.7) 21.8 (17.9, 23.5) 16.9 (15.5, 19.1) White versus Black, p = 0.03 13.3 (10.1, 16.1) 16.7 (15.7, 17.9) p<0.0001 
White versus non-White, p = 0.76 
Cumulative duration of cBTKi exposure, median (95% CI), months 35.1 (32.5, 37.0) 35.3 (31.8, 37.6) 41.7 (34.2, 51.0) 35.6 (32.1, 41.9) White versus Black, p = 0.01 25.1 (17.8, 46.1) 35.9 (33.6, 38.1) p = 0.03 
White versus non-White, p = 0.21 

cBTKi, covalent Bruton tyrosine kinase inhibitor; IQR, interquartile range; CI, confidence interval.

aExcluding patients with missing treatment history, where a specific line of therapy or dates of initiation/discontinuation could not be determined. Non-White includes Black, Asian, and other nonspecified races.

bLog-rank test/Kaplan-Meier method for time to event outcomes of duration of exposure; χ2 test for categorical variables; and t test for continuous variables.

Time to Next Treatment or Death

There were no significant differences in TTNT-D for either race or ethnicity in either adjusted or unadjusted analyses (Table 4). Median time from the start of cBTKi therapy to the start of the next treatment or death was 36.9 months (95% CI: 32.6–41.4) for Black, 35.0 months (95% CI: 33.3–36.9) for White, and 34.1 (95% CI: 29.1–37.8) for non-White patients (all p > 0.05). TTNT-D for those who were Hispanic or Latino was also not significantly different from those who were not Hispanic or Latino (33.9 months, 95% CI: 23.2–49.0 vs. 35.3 months, 95% CI: 33.5–37.1).

Table 4.

Time from the start of cBTKi therapy to the start of next treatment or death

CohortMediana (95% CI), monthsUnadjusted, p valueaAdjusted analysisb, hazard ratio (95% CI), p value
Whitec 35.0 (33.3, 36.9) 0.69 1.02 (0.92, 1.13), p = 0.68 
Non-Whited 34.1 (29.1, 37.8) 
Whitec 35.0 (33.3, 36.9) 0.39 0.95 (0.83, 1.10), p = 0.48 
Black 36.9 (32.6, 41.4) 
Not Hispanic or Latinoc 35.3 (33.5, 37.1) 0.74 0.96 (0.78, 1.19), p = 0.73 
Hispanic or Latino 33.9 (23.2, 49.0) 
CohortMediana (95% CI), monthsUnadjusted, p valueaAdjusted analysisb, hazard ratio (95% CI), p value
Whitec 35.0 (33.3, 36.9) 0.69 1.02 (0.92, 1.13), p = 0.68 
Non-Whited 34.1 (29.1, 37.8) 
Whitec 35.0 (33.3, 36.9) 0.39 0.95 (0.83, 1.10), p = 0.48 
Black 36.9 (32.6, 41.4) 
Not Hispanic or Latinoc 35.3 (33.5, 37.1) 0.74 0.96 (0.78, 1.19), p = 0.73 
Hispanic or Latino 33.9 (23.2, 49.0) 

CI, confidence interval.

aMedian is based on the unadjusted Kaplan-Meier method; p value is based on the log-rank test.

bAdjusted by year of CLL diagnosis, age at diagnosis, sex, race, ethnicity, site of care (community vs. academic practice), socioeconomic status, practice size (total number of patients with CLL treated at the site of care), insurance type, and del(17)(p) status using a Cox proportional hazards regression model; p value is based on the Wald test.

cReference group in the unadjusted and adjusted analyses.

dNon-White includes Black, Asian, and other nonspecified races, excluding those with missing race.

Overall Survival

Survival outcomes by race were not significantly different in either unadjusted or adjusted analyses (Table 5). Median OS from the start of therapy was 79.4 months (95% CI: 70.1 – not evaluable) for Black, 84.1 months (95% CI: 81.1–91.3) for White, 81.7 months (95% CI: 72.3-not evaluable) for non-White patients. Median OS was 89.6 months (95% CI: 70.8–not evaluable) for those who were Hispanic or Latino and 84.1 months (95% CI: 80.5–91.3) for those not Hispanic or Latino. Findings were consistent when evaluating OS from the date of diagnosis, accounting for the time from diagnosis to initiation of treatment in the calculation of total survival time. Survival curves are shown in Figure 3. Factors significantly associated with OS from the start of first-line therapy by race included del 17p, age, sex, year of start of first-line therapy, and site of care (Table 6). Factors not significantly associated with OS by race included socioeconomic status, practice volume, insurance type, and ethnicity. Interactions between ethnicity and site of care and also between race and site of care were not statistically significant.

Table 5.

Overall survival from the start of first-line therapy

CohortMediana (95% CI), monthsUnadjusted, p valueaAdjusted analysisb, hazard ratio (95% CI), p value
Whitec 84.1 (81.1, 91.3) 0.63 1.03 (0.86, 1.23), p = 0.74 
Non-Whited 81.7 (72.3, NE) 
Whitec 84.1 (81.1, 91.3) 0.23 1.04 (0.82, 1.33), p = 0.72 
Black 79.4 (70.1, NE) 
Not Hispanic or Latinoc 84.1 (80.5, 91.3) 0.16 0.63 (0.41, 0.96), p = 0.03 
Hispanic or Latino 89.6 (70.8, NE) 
CohortMediana (95% CI), monthsUnadjusted, p valueaAdjusted analysisb, hazard ratio (95% CI), p value
Whitec 84.1 (81.1, 91.3) 0.63 1.03 (0.86, 1.23), p = 0.74 
Non-Whited 81.7 (72.3, NE) 
Whitec 84.1 (81.1, 91.3) 0.23 1.04 (0.82, 1.33), p = 0.72 
Black 79.4 (70.1, NE) 
Not Hispanic or Latinoc 84.1 (80.5, 91.3) 0.16 0.63 (0.41, 0.96), p = 0.03 
Hispanic or Latino 89.6 (70.8, NE) 

CI, confidence interval; NE, not evaluable.

aMedian is based on the unadjusted Kaplan-Meier method; p value is based on the log-rank test.

bAdjusted by year of CLL diagnosis, age at diagnosis, sex, race, ethnicity, site of care (community vs. academic practice), socioeconomic status, practice size (total number of patients with CLL treated at the site of care), insurance type, and del(17)(p) status using a Cox proportional hazards regression model; p-value is based on the Wald test.

cReference group in the unadjusted and adjusted analyses.

dNon-White includes Black, Asian, and other nonspecified races, excluding those with missing race.

Fig. 3.

Overall survival from diagnosis (left) and start of first-line therapy (right).

Fig. 3.

Overall survival from diagnosis (left) and start of first-line therapy (right).

Close modal
Table 6.

Factors significantly associated with overall survival from the start of first-line therapy in the main analysis, White and Black race

CovariateComparisonAdjusted analysisa, hazard ratio (95% CI), p value
Deletion17p Absent versus present 0.64 (0.54, 0.76), p < 0.0001 
Age 65+ versus 40–65 years 2.73 (2.25, 3,33), p < 0.0001 
Sex Male versus female 1.32 (1.13, 1,54), p = 0.0004 
Site of care Academic/both versus community 0.45 (0.32, 0.64), p < 0.0001 
Year of start of first-line therapyb 2015 versus 2013–14 1.13 (0.89, 1.42), p = 0.32 
2016 versus 2013–14 1.18 (0.93, 1.50), p = 0.17 
2017 versus 2013–14 1.19 (0.93, 1.54), p = 0.17 
2018 versus 2013–14 1.31 (1.00, 1.73), p = 0.05 
2019 versus 2013–14 1.66 (1.23, 2.23), p = 0.0008 
2020 versus 2013–14 1.51 (1.02, 2.22), p = 0.04 
2021 versus 2013–14 1.79 (1.06, 3.03), p = 0.03 
CovariateComparisonAdjusted analysisa, hazard ratio (95% CI), p value
Deletion17p Absent versus present 0.64 (0.54, 0.76), p < 0.0001 
Age 65+ versus 40–65 years 2.73 (2.25, 3,33), p < 0.0001 
Sex Male versus female 1.32 (1.13, 1,54), p = 0.0004 
Site of care Academic/both versus community 0.45 (0.32, 0.64), p < 0.0001 
Year of start of first-line therapyb 2015 versus 2013–14 1.13 (0.89, 1.42), p = 0.32 
2016 versus 2013–14 1.18 (0.93, 1.50), p = 0.17 
2017 versus 2013–14 1.19 (0.93, 1.54), p = 0.17 
2018 versus 2013–14 1.31 (1.00, 1.73), p = 0.05 
2019 versus 2013–14 1.66 (1.23, 2.23), p = 0.0008 
2020 versus 2013–14 1.51 (1.02, 2.22), p = 0.04 
2021 versus 2013–14 1.79 (1.06, 3.03), p = 0.03 

aAdjusted by year of CLL diagnosis, age at diagnosis, sex, race, ethnicity, site of care (community vs. academic practice), socioeconomic status, practice size (total number of patients with CLL treated at the site of care), insurance type, and deletion17p status using a Cox proportional hazards regression model; p value is based on the Wald test.

bCensoring rates increase as the year of start of first-line therapy advances; data related to this variable should be interpreted with caution.

CLL (inclusive of SLL) is a heterogeneous hematological malignancy, characterized by different molecular subtypes that have historically been associated with worse survival outcomes. Unmutated IGHV status, for instance, is associated with shorter treatment-free interval as well as inferior survival, irrespective of stage at the time of diagnosis [13, 14]. Del(11q) has been associated with a more symptomatic presentation at the time of diagnosis, including B symptoms and extensive lymphadenopathy, possibly contributing to earlier treatment initiation [15]. On the other hand, del(13q) alterations are associated with a much more indolent biological behavior and longer survival [15].

Black patients have been described as younger and portending more symptoms upon initial diagnosis and, in some analysis, twice more likely to die from CLL in the first 5 years of diagnosis than their White counterparts [16, 17]. A retrospective analysis of CLL cases seen at 2 different academic institutions demonstrated higher rates of molecular alterations associated with worse outcomes such as del(17p) and del(11q) and unmutated IGVH gene among individuals of Black race, as well as worse OS than the non-Black population [18]. Similarly, this study demonstrates differences in molecular subtypes of CLL that may explain aggressive biological behavior of this disease in Black patients that may have contributed to potentially worse outcomes observed in these earlier studies.

While data have shown racial disparities for patients with CLL in the USA over the past 2 decades, these studies do not assess the treatments received or other factors that could be related to survival outcomes after the introduction of targeted therapies. A retrospective analysis of 47,867 cases diagnosed from 2006 to 2016 from the SEER 18 database demonstrated Black race to be associated with worse OS (HR = 1.5, 95% CI: 1.4–1.6), remaining the strongest independent factor for OS after controlling for SES [19]. An analysis of the National Cancer Database (NCDB) of 97,804 CLL cases diagnosed between 2004 and 2018 also demonstrated a relationship with Black race and shorter OS compared to their White counterparts (HR = 1.5, 95% CI: 1.5–1.6) [3]. Neither study describes treatment patterns nor exposure to cBTKi therapy as the majority of the time period studied was in the pre-cBTKi era. In the current study of 6,372 patients diagnosed with CLL from 2013 to 2022, patients of Black race displayed higher rates of unmutated IGHV status and del(11q), presented at younger age and at more advanced stage than White patients. They were, however, more likely to be treated with a cBTKi than White patients, despite lower SES. Similar to the current study, an analysis of 1,459 patients captured by the Inform CLL registry treated with cBTKi from 2015 to 2019 showed that there were no racial disparities in survival despite Black patients presenting at younger age, with a higher Rai stage and a worse ECOG performance status, underscoring the potential of access to effective therapy to overcome unfavorable outcomes [9]. The findings of the Inform CLL registry, consistent with the current study, suggest that the racial disparity in survival may have been reduced in recent years. Furthermore, when including the time period of diagnosis to treatment in the duration of OS, the results remain unchanged, suggesting that there are not delays in time to treatment that could have impacted the OS findings observed. Importantly, this study is limited to the treated population, and OS findings cannot be extrapolated to patients with CLL who do not receive systemic therapy.

The introduction of cBTKi therapy has revolutionized the treatment of CLL, especially patients without TP53 mutations/del(17p). Multiple trials have demonstrated the efficacy of cBTKi therapy versus cytotoxic chemotherapy [20‒22]. However, access to oral agents may be restricted to certain patients due to lack of insurance coverage, potentially generating disparities in outcomes. These data support the emerging evidence, suggesting that the outcome disparities seen before the cBTKi era may no longer be present and leads to hypotheses about the causal nature of this relationship that should be further investigated. Causal inference regarding cBTKi therapy or the timing of when this therapy is received cannot be made in the current study. The introduction of BCL2i therapy and its use in sequence with both covalent and noncovalent BTKi therapy could also be evaluated in future research, once sufficient follow-up is available for patients with access to all these agents. Prospective trials or retrospective analyses incorporating time-varying confounders would be needed to infer causality. Therefore, this study, while demonstrating the association of these variables, cannot conclude that the use of a targeted or other novel therapy is directly responsible for the outcomes observed.

While the results from this study demonstrate similar survival outcomes by race, additional data are needed to confirm this conclusion. Patients who are Black continue to present with clinical characteristics associated with poor prognosis (e.g., younger age, unmutated IGHV, more advanced stage of disease) as in the pre-cBTKi era, yet the current data show that these patients now receive faster treatment and are more likely to receive BTKi-based therapy than those who are White. This study did not specifically look at time-to-treatment outcomes, nor was there access to data from the pre-diagnosis period, to identify reasons for the later stage at diagnosis. The potential gaps in care leading to a later-stage diagnosis as well as rates of treatment are also important topics of future investigation, as patients who did not receive systemic therapy are not recorded in this dataset. These promising data demonstrate the potential impact of reducing barriers to novel treatments and emphasize the need to continue to reduce barriers for all patients. However, in adjusted analyses, there were differences in OS related to ethnicity; the reasons for this finding remain unclear, as sample sizes were small and the follow-up was limited. This outcome deserves more attention in a larger cohort with a longer follow-up. Though this study specifically evaluated race and ethnicity, these data are limited with regard to other races. For example, there are no data recorded for Native American or Hawaiian populations, and the Asian population sample size was too small to report without risking patient identification. These issues demonstrate the gap in understanding about other racial groups that are diagnosed with CLL. Underrepresented populations in clinical research supporting these novel agents are not limited to racial or ethnic groups. The care and outcomes of patients treated in settings without access to clinical trials, patients who are uninsured or underinsured, as well those who reside in rural regions for example, remain largely unknown. Additional work and expansion of data that include more robust social determinants of health data are needed to ensure equity in the care of all patients with CLL.

Patients who are Black tend to present with characteristics suggesting more aggressive disease at diagnosis of CLL as compared to White patients. Although these findings have been consistent through time and warrant further study to understand and address root causes, recent analyses have shown that fortunately progress has been made in closing the survival gap. Studies such as this one conducted in the post-cBTKi era have demonstrated that Black patients do not have significantly different survival outcomes as those who are White. Furthermore, and interestingly, among all treated patients, those who are Black are more likely to initiate therapy earlier and more likely to receive cBTKi therapy for CLL than White patients.

De-identified data are not considered human subjects in accordance with US 45 CFR 46.

Drs. Hess, Winfree, and Escalon and Ms. Li are employees of Eli Lilly and Company.

This was an unfunded research project supported by employee time and database access provided by Eli Lilly and Company.

Debora Bruno: conceptualization; investigation; and writing – review and editing. Manoj Khanal and Xiaohong Ivy Li: formal analysis, validation, data curation, and writing – review and editing. Mercer Escalon: conceptualization and writing – review and editing. Katherine Winfree: investigation and writing – review and editing. Lisa Hess: conceptualization, methodology, validation, investigation, writing – original draft, and visualization.

The data that support the findings of this study were originated by and are the property of Flatiron Health, Inc., which has restrictions prohibiting the authors from making the dataset publicly available. Requests for data sharing by license or by permission for the specific purpose of replicating results in this manuscript can be submitted to [email protected].

1.
SEER
.
SEER*Explorer: an interactive website for SEER cancer statistics
.
2023
; Available from: https://seer.cancer.gov/statistics-network/explorer/. Data source(s): SEER Incidence Data, November 2022 Submission (1975-2020)
2.
Casey
M
,
Odhiambo
L
,
Aggarwal
N
,
Shoukier
M
,
Garner
J
,
Islam
KM
, et al
.
Are pivotal clinical trials for drugs approved for leukemias and multiple myeloma representative of the population at risk
.
J Clin Oncol
.
2022
;
40
(
32
):
3719
29
.
3.
Vardell
V
,
Ermann
DA
,
Shah
H
,
Fitzgerald
L
,
Hu
B
,
Stephens
DM
.
Influence of ethnic idenity on overall survival in patients with chronic lymphocytic leukemia
.
Am J Hematol
.
2023
;
98
(
7
):
E172
4
.
4.
Young
RM
,
Staudt
LM
.
Targeting pathological B cell receptor signalling in lymphoid malignancies
.
Nat Rev Drug Discov
.
2013
;
12
(
3
):
229
43
.
5.
Sun
Y
,
Zhao
X
,
Ding
N
,
Gao
H
,
Wu
Y
,
Yang
Y
, et al
.
PROTAC-induced BTK degradation as a novel therapy for mutated BTK C481S induced ibrutinib-resistant B-cell malignancies
.
Cell Res
.
2018
;
28
(
7
):
779
81
.
6.
NCCN
.
NCCN clinical practice guidelines in Oncology: chronic lymphocytic leukemia/small lymphocytic leukemia
.
2023
. Version 3.2023. Available from: www.nccn.org
7.
Fischer
L
.
FDA grants pirtobrutinib accelerated approval to treat CLL/SLL in the third line
.
Oncol Nurs News
.
2023
. NA-NA.
8.
Deeks
ED
.
Venetoclax: first global approval
.
Drugs
.
2016
;
76
(
9
):
979
87
.
9.
Barrientos
JC
.
Real-world outcomes by race in patients (pts) with chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL): final analysis results from the informCLL registry
.
American Society of Clinical Oncology
;
2023
.
10.
Ma
X
.
Comparison of population characteristics in real-world clinical Oncology databases in the US: flatiron health, SEER, and NPCR
.
medRxiv
.
2020
. 2020.03.16.20037143.
11.
Birnbaum
B
.
Model-assisted cohort selection with bias analysis for generating large-scale cohorts from the EHR for oncology research
.
2020
. arXiv preprint arXiv:2001.09765.
12.
NIH
.
Definition of human subjects research
.
2024
. Available from: https://grants.nih.gov/policy/humansubjects/research.htm
13.
Damle
RN
,
Wasil
T
,
Fais
F
,
Ghiotto
F
,
Valetto
A
,
Allen
SL
, et al
.
IgV gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia
.
Blood
.
1999
;
94
(
6
):
1840
7
.
14.
Oscier
DG
,
Gardiner
AC
,
Mould
SJ
,
Glide
S
,
Davis
ZA
,
Ibbotson
RE
, et al
.
Multivariate analysis of prognostic factors in CLL: clinical stage, IGVH gene mutational status, and loss or mutation of the p53 gene are independent prognostic factors
.
Blood
.
2002
;
100
(
4
):
1177
84
.
15.
Döhner
H
,
Stilgenbauer
S
,
Benner
A
,
Leupolt
E
,
Kröber
A
,
Bullinger
L
, et al
.
Genomic aberrations and survival in chronic lymphocytic leukemia
.
N Engl J Med
.
2000
;
343
(
26
):
1910
6
.
16.
Shenoy
PJ
,
Malik
N
,
Sinha
R
,
Nooka
A
,
Nastoupil
LJ
,
Smith
M
, et al
.
Racial differences in the presentation and outcomes of chronic lymphocytic leukemia and variants in the United States
.
Clin Lymphoma Myeloma Leuk
.
2011
;
11
(
6
):
498
506
.
17.
Coombs
CC
,
Falchi
L
,
Weinberg
JB
,
Ferrajoli
A
,
Lanasa
MC
.
Chronic lymphocytic leukemia in African Americans
.
Leuk Lymphoma
.
2012
;
53
(
11
):
2326
9
.
18.
Falchi
L
,
Keating
MJ
,
Wang
X
,
Coombs
CC
,
Lanasa
MC
,
Strom
S
, et al
.
Clinical characteristics, response to therapy, and survival of African American patients diagnosed with chronic lymphocytic leukemia: joint experience of the MD Anderson Cancer Center and Duke University Medical Center
.
Cancer
.
2013
;
119
(
17
):
3177
85
.
19.
Kittai
A
.
Racial and socioeconomic disparities among patients with chronic lymphocytic leukemia: analysis of Surveillance, Epidemiology, and End Results program data
.
American Society of Clinical Oncology
;
2022
.
20.
Burger
JA
,
Barr
PM
,
Robak
T
,
Owen
C
,
Ghia
P
,
Tedeschi
A
, et al
.
Long-term efficacy and safety of first-line ibrutinib treatment for patients with CLL/SLL: 5 years of follow-up from the phase 3 RESONATE-2 study
.
Leukemia
.
2020
;
34
(
3
):
787
98
.
21.
Sharman
JP
,
Egyed
M
,
Jurczak
W
,
Skarbnik
A
,
Pagel
JM
,
Flinn
IW
, et al
.
Efficacy and safety in a 4-year follow-up of the ELEVATE-TN study comparing acalabrutinib with or without obinutuzumab versus obinutuzumab plus chlorambucil in treatment-naïve chronic lymphocytic leukemia
.
Leukemia
.
2022
;
36
(
4
):
1171
5
.
22.
Munir
T
,
Shadman
M
,
Robak
T
,
R. Brown
J
,
Kahl
B
,
Ghia
P
, et al
.
P639: zanubrutinib (ZANU) vs Bendamustine+ Rituximab (BR) in patients (pts) with treatment-naïve chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL): extended follow-up of the SEQUOIA study
.
Hemasphere
.
2023
;
7
(
S3
):
e15364af
.