3,036
Views
3
CrossRef citations to date
0
Altmetric
Articles

Real-world healthcare resource utilization and costs associated with tisagenlecleucel and axicabtagene ciloleucel among patients with diffuse large B-cell lymphoma: an analysis of hospital data in the United States

, , , , , , ORCID Icon, & show all
Pages 2052-2062 | Received 07 Jan 2022, Accepted 22 Mar 2022, Published online: 14 Apr 2022

Abstract

This study compared the real-world healthcare resource utilization (HRU), costs, adverse events (AEs), and AE treatments associated with the chimeric antigen receptor T-cell (CAR-T) therapies, tisagenlecleucel (tisa-cel) and axicabtagene ciloleucel (axi-cel), for relapsed/refractory diffuse large B-cell lymphoma (r/r DLBCL). Adults with DLBCL who received tisa-cel or axi-cel were identified in the Premier Healthcare Database (2017–2020). Non-CAR-T costs, HRU, and AE rates during the infusion and follow-up periods were compared between the tisa-cel and axi-cel cohorts. Of 119 patients, 33 received tisa-cel (86% as inpatient infusion) and 86 received axi-cel (100% inpatient). Tisa-cel was associated with significantly shorter mean inpatient length of stay than axi-cel during infusion (11.3 vs. 18.3 days) and follow-up ([monthly] 3.9 vs. 6.9 days). Non-CAR-T costs were significantly lower for tisa-cel compared with axi-cel during infusion ($27594.8 vs. $51378.3) and follow-up ([monthly] $28777.3 vs. $46575.7; both p< .05). Rates of AEs and AE treatments were similar.

Introduction

In the United States (US), diffuse large B-cell lymphoma (DLBCL) has an estimated annual incidence of 5.6 per 100,000 people that increases with age; the median age at presentation is 70 years [Citation1–3]. Approximately 30–40% of patients with DLBCL relapse after first-line treatment or demonstrate refractory disease [Citation4,Citation5], which are associated with a worse prognosis [Citation4,Citation5]. For relapsed or refractory (r/r) DLBCL, rituximab-based chemotherapy followed by autologous hematopoietic cell transplantation (HCT) provides the best chance of cure, although only half of patients are eligible for this approach [Citation6]. Among those eligible, another half eventually relapse following autologous HCT [Citation7–13], leaving historically few therapeutic options before the advent of chimeric antigen receptor T-cell (CAR-T) therapy for relapsed/refractory diffuse large B-cell lymphoma (r/r DLBCL) [Citation5,Citation12,Citation14–16].

CAR-T therapies engineer lymphoma patients’ extracted T-cells to target the CD19 antigen expressed by both normal and malignant B-cells after reinfusion into the patient [Citation9,Citation17–19]. Three CAR-T therapies have been approved by the US Food and Drug Administration for r/r DLBCL: axicabtagene ciloleucel (axi-cel) in 2017, tisagenlecleucel (tisa-cel) in 2018, and lisocabtagene maraleucel in 2021. These therapies were approved based on the efficacy demonstrated in their individual, single-arm clinical trials of ZUMA-1, JULIET, and TRANSCEND, respectively [Citation9,Citation17–19].

While the clinical benefits of CAR-T therapies have been well-established, few studies have compared the real-world healthcare resource utilization (HRU), costs, and adverse events (AEs) associated with CAR-T therapy for r/r DLBCL in the US. As axi-cel and tisa-cel have now been approved in the US for r/r DLBCL since 2017 and 2018, respectively, there are sufficient data for assessments of real-world economic and safety outcomes. However, most prior real-world studies have used data collected in registries or medical centers, which typically do not focus on costs, while others using claims, hospital-based databases, or public data have not differentiated between CAR-T products [Citation20–22]. Additionally, some studies have examined just the healthcare costs [Citation23,Citation24] or HRU [Citation20,Citation25] associated with axi-cel and/or tisa-cel, but rarely both or comparatively. To address this knowledge gap, this study assessed and compared the real-world HRU and costs incurred by US hospitals, as well as treatment-related AEs, associated with axi-cel or tisa-cel for r/r DLBCL using data from the Premier Healthcare Database.

Methods

Data source

This retrospective non-interventional cohort study used data acquired from the Premier Healthcare Database (2017 Q1–2020 Q1), which contains health records from >1000 contributing US hospitals and affiliated outpatient facilities, including data from approximately 231 million unique patients. It encompasses demographic and disease diagnoses, payer/financial information, and claims of inpatient admissions, outpatient visits, lab tests, and pharmacy services but lacks detailed disease-associated data. As the data used in this study were deidentified, no Institutional Review Board oversight was required.

Study population

Adults (aged ≥18 years) with ≥1 primary or secondary diagnosis of DLBCL (International Classification of Diseases, 10th edition, Clinical Modification [ICD-10 CM] code: C83.3x) and who received the CAR-T therapies tisa-cel or axi-cel following DLBCL diagnosis were included (). Patients with DLBCL but without associated identifiable CAR-T product names were excluded. Patients were classified into tisa-cel and axi-cel cohorts depending on the CAR-T product received. Although patients receiving tisa-cel or axi-cel were assumed to have r/r DLBCL per the CAR-T prescribing labels at the time of the study [Citation26,Citation27], r/r status was not clearly defined and available in the database.

Figure 1. Sample selection flowchart. axi-cel: axicabtagene ciloleucel; CAR-T: chimeric antigen receptor modified T-cells; DLBCL: diffuse large B-cell lymphoma; Q: quarter; tisa-cel: tisagenlecleucel.

Figure 1. Sample selection flowchart. axi-cel: axicabtagene ciloleucel; CAR-T: chimeric antigen receptor modified T-cells; DLBCL: diffuse large B-cell lymphoma; Q: quarter; tisa-cel: tisagenlecleucel.

Study design

The index date was the admission date for the CAR-T infusion encounter for inpatient infusion or the infusion encounter date for outpatient infusion. The infusion encounter was the hospitalization stay associated with inpatient infusion or the outpatient visit for outpatient infusion. The study period spanned from the index date to the end of data availability.

Study measures

Baseline characteristics assessed at the index date included demographics (age, sex, race); disease characteristics (National Cancer Institute Comorbidity Index [NCICI] and individual comorbidities); treatment characteristics (index year, physician specialty); and hospital characteristics (provider region, urbanicity, teaching facility, and number of beds).

HRU assessed during the study period included inpatient admissions, inpatient length of stay (LOS), intensive care unit (ICU) stays, ICU days, outpatient visits, and readmission within 30 or 60 days after discharge from the infusion admission. The number of visits and LOS per patient per month were calculated as the total number of visits or days/total person-months during the follow-up. Inpatient LOS included hospital and ICU days. Total non-CAR-T healthcare costs during the study period were assessed, defined as internal hospital costs for providing inpatient and outpatient services (i.e. the amount reported by hospitals vs. charges or payment/reimbursement amounts [adjudicated claims]), including tocilizumab [Citation28]. A subgroup analysis was conducted comparing HRU and costs among patients who received inpatient tisa-cel vs. axi-cel.

AEs assessed during the study period included the rate and grade of cytokine release syndrome (CRS), rate of neurological toxicity (NT), rate of hypogammaglobulinemia, and AE-related treatments (i.e. tocilizumab, steroids, and intravenous immunoglobulin [IVIG]). CRS events (grade ≥3) were identified using billing data that indicated ICU and vasopressor or ventilation support within one-month post CAR-T infusion. NT events were identified using ICD-10-CM codes within one-month post CAR-T infusion, and hypogammaglobulinemia events were identified using ICD-10-CM code or the use of IVIG (Supplemental Table 1).

Statistical analysis

Baseline characteristics, and the AEs mentioned above and related treatments, were described and compared between cohorts using Wilcoxon’s rank-sum tests for continuous variables and Chi-square tests for categorical variables. HRU and costs were described and compared between cohorts during two periods: (1) per patient during the infusion encounter and (2) per patient per month over the entire study period (from the infusion encounter to the last observed medical visit within the same facility). HRU was compared using generalized linear models (GLMs) with a Poisson distribution, with an offset to account for varying follow-up times between patients. Unadjusted incidence rate ratios (IRRs) with their 95% confidence intervals (CIs) and p values were reported comparing the tisa-cel and axi-cel cohorts. Costs (in 2020 US dollars) were compared between cohorts using GLMs with a Tweedie distribution and log-link function. Analyses were conducted in SAS® version 9.4 (SAS Institute Inc., Cary, NC). A p< .05 was used to determine significance.

Results

Sample selection

A total of 119 adults with DLBCL who received study-specific CAR-T therapy were identified from 15 medical centers, including 33 in the tisa-cel cohort and 86 in the axi-cel cohort ().

Baseline characteristics

Patient characteristics

Demographics were similar between the cohorts (). The mean ages were 62.3 and 59.8 years for the tisa-cel and axi-cel cohorts, respectively. Over half of patients in both cohorts were male (54.6% [tisa-cel] and 67.4% [axi-cel]) and White (75.8% and 62.8%, respectively). Comorbidities were generally comparable between the cohorts, though few conditions were more prevalent in tisa-cel than axi-cel: hypertension (57.6% vs. 45.4%), diabetes (30.3% vs. 20.9%), and renal disease (21.2% vs. 8.1%), and significantly more patients with depression (36.4% vs. 18.6%, p= .04). The mean NCICI scores were comparable for the tisa-cel (1.6 [standard deviation (SD): 1.8]) and axi-cel (1.4 [1.8]; p= .55) cohorts.

Table 1. Patient baseline characteristics.

Hospital characteristics

Patients were treated in 15 hospital centers (seven for tisa-cel and 11 for axi-cel, with three providing both) (). All centers were in urban areas, and were primarily teaching hospitals (100.0% [tisa-cel] and 81.8% [axi-cel]) and large hospitals with >500 beds (73.3% of all centers).

Table 2. Infusion center characteristics.

Comparison of HRU

HRU during the infusion encounter

During the infusion encounter, 75.8% of the tisa-cel cohort and 100% of the axi-cel cohort received inpatient infusion; among all patients who received infusion, the mean LOS were 11.3 and 18.3 days, respectively (both p< .01) (). The remaining 24.2% of patients in the tisa-cel cohort received outpatient infusion, reported from three centers. Numerically fewer tisa-cel patients were transferred to the ICU during the infusion encounter compared with axi-cel patients (9.1% vs. 20.9%, respectively; p= .13). Among all patients, the mean ICU days were 0.5 and 1.7 days for the tisa-cel and axi-cel cohorts, respectively (p= .14). Among patients with an ICU stay, the mean ICU days were 5.3 and 8.0 days for the tisa-cel and axi-cel cohorts, respectively (p= .92). The post-infusion readmission rates within 30 days (16.0% [tisa-cel] vs. 18.6% [axi-cel]) and 60 days (24.0% vs. 24.4%, respectively) of infusion were comparable.

Table 3. HRU associated with tisa-cel and axi-cel.

HRU during the entire follow-up period

The average length of follow-up after infusion was 5.0 months for the tisa-cel cohort and 3.3 months for the axi-cel cohort (). Despite the longer follow-up for the tisa-cel cohort, a significantly lower proportion had ≥1 inpatient admission during the study period vs. the axi-cel cohort (90.9% vs. 100.0%, respectively; p< .05) as all were admitted for infusion. The remaining 9.1% of tisa-cel patients received outpatient infusion and were not admitted to a hospital during follow-up. The monthly inpatient admission rates during the follow-up period were similar between cohorts (0.33 [tisa-cel] and 0.55 [axi-cel]; IRR [95% CI]: 0.72 [0.47, 1.09]; p= .12). However, the monthly inpatient LOS was significantly shorter for the tisa-cel (3.9 days) compared with the axi-cel cohort (6.8 days; IRR [95% CI]: 0.56 [0.33, 0.96]; p= .03). The mean number of ICU stays per month during follow-up were comparable between cohorts (0.04 [tisa-cel] and 0.09 [axi-cel]; IRR [95% CI]: 0.45 [0.16, 1.29]; p= .14). The mean number of ICU days per month were also similar (0.4 [tisa-cel] and 0.8 [axi-cel] days; 0.51 [0.13, 2.0]; p= .34). The tisa-cel cohort had a significantly higher proportion with ≥1 outpatient visit during follow-up compared with the axi-cel cohort (81.8% vs. 52.3%, respectively; p< .01), as well as a significantly higher rate of outpatient visits per month (2.2 vs. 1.2; IRR [95% CI]: 1.86 [1.13, 3.05]; p= .01).

Comparison of healthcare costs

Costs during the infusion encounter

The total non-CAR-T costs during the infusion encounter were significantly lower for patients receiving tisa-cel compared with axi-cel ($27,595 [SD: $27,995] vs. $51,378 [$38,529], respectively), primarily driven by lower inpatient costs ($26,446 [$28,967] vs. $51,378 [$38,529]; both p< .01) (). The mean non-CAR-T outpatient costs associated with the infusion encounter were $1149 [SD: $2695] for tisa-cel and $0 for axi-cel (no patients received outpatient axi-cel).

Figure 2. Healthcare costs in the tisa-cel and axi-cel cohorts. During both the infusion encounter and over the entire follow-up period, tisa-cel was associated with significantly lower overall costs and inpatient costs compared with axi-cel (all comparisons p< .05). axi-cel: axicabtagene ciloleucel; IP: inpatient; OP: outpatient; PPPM: per-patient-per-month; tisa-cel: tisagenlecleucel.

Figure 2. Healthcare costs in the tisa-cel and axi-cel cohorts. During both the infusion encounter and over the entire follow-up period, tisa-cel was associated with significantly lower overall costs and inpatient costs compared with axi-cel (all comparisons p< .05). axi-cel: axicabtagene ciloleucel; IP: inpatient; OP: outpatient; PPPM: per-patient-per-month; tisa-cel: tisagenlecleucel.

Costs during the entire follow-up period

During the entire follow-up period, monthly non-CAR-T costs were significantly lower for the tisa-cel cohort compared with the axi-cel cohort ($28,777 [SD: $32,852] vs. $46,576 [$39,140], respectively; p< .05), driven by lower monthly inpatient costs ($23,844 [$32,160] vs. $44,561 [$39,657]; p< .01) (). The monthly outpatient costs were significantly higher for the tisa-cel cohort compared with the axi-cel cohort ($4933 [SD: $7950] vs. $2014 [$6117], respectively; p< .01).

Subgroup analysis of HRU and healthcare costs (inpatient infusion)

The results of the subgroup analysis comparing HRU (Supplemental Table 2) and non-CAR-T costs (Supplemental Table 3) between patients who received inpatient tisa-cel (n = 25) vs. axi-cel (n = 86) were directionally consistent with the main analyses, but most comparisons were not significantly different due to the reduction in sample size. However, the inpatient costs at the infusion encounter remained significantly higher for axi-cel vs. tisa-cel ($51,378 [SD: $38,528] vs. $34,908 [$28,479]; p< .01).

Comparison of AEs and related treatments

CRS and related treatments

Within the one-month period post-infusion, 6.1% (n = 2) of the tisa-cel and 15.1% (n = 13) of the axi-cel cohorts experienced grade ≥3 CRS (p= .23) (). A significantly lower proportion of the tisa-cel cohort compared with axi-cel cohort used tocilizumab (15.2% vs. 51.2%, respectively; p< .01); of these patients, the number of tocilizumab infusions were comparable between cohorts (1.2 vs. 1.9 infusions; p= .16).

Table 4. AEs and related treatments in the tisa-cel and axi-cel cohorts.

NT and steroid use

A numerically lower proportion of the tisa-cel cohort experienced NT events than the axi-cel cohort (21.2% [n = 7] vs. 36.0% [n = 31]; p= .12) (). The most common NT event among both cohorts was encephalopathy (15.2% [tisa-cel] and 29.1% [axi-cel]; p= .12). Among patients with NT events in both cohorts, the proportions with steroid use (71.4% [tisa-cel] vs. 77.4% [axi-cel]; p> .99) and the number of days on steroids among users (12.0 vs. 12.5 days, respectively; p= .93) were similar.

Hypogammaglobulinemia and IVIG use

The tisa-cel cohort had a numerically higher proportion of patients with hypogammaglobulinemia compared with the axi-cel cohort (30.3% vs. 19.8%, respectively; p= .22) (). Among patients with hypogammaglobulinemia in both cohorts, the proportion with IVIG use (80.0% [tisa-cel] vs. 88.2% [axi-cel]; p = .61) and the number of days on IVIG among users (3.0 vs. 2.1 days, respectively; p= .23) were comparable, although the tisa-cel cohort had a longer follow-up time.

Discussion

This study compared the real-world HRU, healthcare costs, and select AEs associated with tisa-cel and axi-cel for r/r DLBCL using a large US hospital database. To our knowledge, it is the first study to assess HRU and provider costs associated with tisa-cel and axi-cel in a real-world setting during both the infusion encounter and the period following (and including) infusion. Patient characteristics were generally similar between cohorts, although tisa-cel patients had higher rates of some comorbidities, similar to observations of prior studies [Citation29–32]. Our sample was slightly younger than patients in the Center for International Blood & Marrow Transplant Research (CIBMTR) registry (mean age: 59.8–62.3 vs. 62.0–65.4 years, respectively) [Citation30,Citation32].

The results of the main analysis indicated that, compared with axi-cel, tisa-cel was associated with significantly fewer inpatient admissions and significantly shorter LOS during both the infusion and follow-up periods. Lower inpatient HRU associated with tisa-cel vs. axi-cel led to substantially lower non-CAR-T-related healthcare costs during both the infusion encounter and follow-up period. During the follow-up period, the tisa-cel cohort had a significantly lower proportion with ≥1 inpatient admission but significantly more outpatient visits vs. the axi-cel cohort. Directionally similar findings were observed in the subgroup analyses comparing costs and HRU among patients who received inpatient tisa-cel or axi-cel infusion, although due to the small sample sizes, statistical significance was largely lost, and those results should be interpreted with caution.

Approximately, one-fourth of the tisa-cel cohort received outpatient infusion while the entire axi-cel cohort received inpatient infusion, consistent with prior real-world studies reporting that tisa-cel is more likely to be administered outpatient vs. axi-cel [Citation29,Citation33]. This difference is likely influenced by the expected onset of AEs like CRS and the treatment administration settings of the pivotal CAR-T trials [Citation22,Citation34]. Some patients in JULIET received outpatient tisa-cel infusion [Citation9], permitting a safety/feasibility evaluation, while no patients in ZUMA-1 received outpatient axi-cel infusion [Citation19]. Additionally, axi-cel has shorter onset and greater severity of CRS vs. tisa-cel, which affects the suitability for outpatient administration [Citation9,Citation18,Citation19]. This observation is consistent with the current finding that tisa-cel was associated with numerically lower rates of grade ≥3 CRS and NT and significantly lower use of tocilizumab than axi-cel. Similarly, the generally higher utilization of inpatient HRU among the axi-cel cohort reported within this study reflects requirements that axi-cel patients be monitored for CRS and NT for ≥7 days following infusion [Citation26]. As outpatient CAR-T therapy infusion becomes more common with increased clinician experience and the development of CRS prophylaxis strategies, future analyses should be conducted when new data emerge.

Two prior studies have reported inpatient LOS for the CAR-T infusion encounter of 15 and 19.1 days using the Vizient Hospital Database and 100% Medicare data, respectively [Citation21,Citation35], but did not differentiate tisa-cel from axi-cel. In the present study, the monthly inpatient LOS over the follow-up period was significantly shorter for the tisa-cel vs. the axi-cel cohort (3.9 vs. 6.8 days), as was the mean LOS during the infusion encounter (11.3 vs. 18.3 days; both p< .05). Among patients who received inpatient infusion, the monthly inpatient LOS was numerically shorter for the tisa-cel vs. the axi-cel cohort (4.5 vs. 6.8 days). These findings are directionally consistent with a 2020 chart review study by Riedell et al. [Citation29] which reported a median LOS of 15 days for axi-cel (92% inpatient infusion) and two days for tisa-cel (36% inpatient infusion) within 28-days post-infusion. As the majority of tisa-cel patients in that study received outpatient infusion, the median was mainly driven by those treated in that setting.

The non-CAR-T costs associated with tisa-cel and axi-cel infusion observed in this study were not trivial. The tisa-cel cohort incurred significantly lower mean costs than the axi-cel cohort during the infusion encounter ($27,595 vs. $51,378, respectively) due to significantly lower mean inpatient costs ($26,446 vs. $51,378). Among patients who received inpatient infusion, the tisa-cel cohort also incurred significantly lower mean costs during the infusion encounter than axi-cel ($34,909 vs. $51,378, respectively). Two studies by Harris et al. [Citation35,Citation36] using the Vizient Database and cost-to-charge ratios, reported median costs of $27,189 to $37,763 for inpatient CAR-T infusion encounters (excluding CAR-T product cost) from a provider's perspective. The costs in those studies were not separated by therapy but still reflect an overlapping range with the present results. Maziarz et al. found that during the period spanning from lymphodepleting chemotherapy to day 30 post-CAR-T infusion, patients receiving tisa-cel vs. axi-cel inpatient infusion incurred lower mean hospital charges (excluding CAR-T costs) ($174,782 vs. $198,209) [Citation37]. As real-world experience with CAR-T infusion settings grows, future studies should consider logistical issues, direct costs, and indirect costs (e.g. lost productivity, cost of transportation) for patients and caregivers [Citation34].

The present rates of grade ≥3 CRS (6.1% [tisa-cel] vs. 15.1% [axi-cel]) were lower than those reported for tisa-cel in JULIET (22%, using the American Society for Transplantation and Cellular Therapy [ASTCT] grading criteria) but similar to those reported for axi-cel in ZUMA-1 (13%, using the Lee grading criteria) [Citation9,Citation19]. However, clinical practice related to CAR-T therapy and AE management have evolved since those trials’ conduct, which is reflected in the lower real-world rates of grade ≥3 CRS. Two studies by Pasquini et al., using CIBMTR registry data, reported a grade ≥3 CRS rate of 4.5% with tisa-cel (ASTCT criteria) [Citation30] and 8–10% with axi-cel (Lee criteria) [Citation31]. Similarly, another CIBMTR study by Jaglowski et al. reported a grade ≥3 CRS rate of 4% for tisa-cel [Citation38]. A 2021 study by Riedell et al. of outcomes at seven CAR-T infusion centers reported grade ≥3 CRS rates of 2% for tisa-cel and 9% for axi-cel (ASTCT criteria) [Citation39]. Our CRS identification was based on billing data, incorporating the ASTCT algorithm and clinical input, and demonstrated high specificity. Of the 15 patients identified as having CRS via billing data, 14 received tocilizumab during the same visit as CRS was recorded; the remaining patient received axi-cel and was classified as having grade 5 CRS due to an in-hospital death. The currently reported rates of tocilizumab use are also consistent with those of recent real-world studies: 15% and 61% for tisa-cel and axi-cel, respectively, in Riedell et al. [Citation39] and 67% for axi-cel in Jacobson et al. [Citation32].

This study is subject to several limitations, some of which are inherent to retrospective analyses of hospital databases (e.g. miscoding and missing data). First, the Premier data are mainly inpatient, which may underrepresent outpatient CAR-T infusions. Second, the cohort sample sizes were generally small, especially for tisa-cel, which limits the statistical power to identify significant differences between cohorts. Third, due to the limited sample size and because the majority of baseline patient and hospital characteristics were comparable between the cohorts, the HRU and costs comparisons were conducted without adjusting for potential baseline differences or center-specific factors. Fourth, due to the lack of clinical data, treatments prior to CAR-T infusion and DLBCL staging were not assessed in this study. In the CIBMTR registry, tisa-cel patients (median age: 65.4 years) had a median of four prior therapies, 26% had prior autologous HCT, and ∼95% had primary r/r DLBCL; axi-cel patients were slightly younger (median: 62 years), 29% had prior autologous HCT, and 66% had chemotherapy-resistant disease prior to infusion [Citation30,Citation32]. Overall survival could not be assessed due to the lack of detailed death information in the database. In addition, CRS was identified based on resource use while NT events were identified from ICD-10-CM codes rather than the ASTCT criteria with grading system. This may have under- or overestimated some AEs and the results may be less comparable with prior studies using detailed clinical data. For example, steroids may be used for conditions other than CAR-T AEs, but the database did not detail the prescribed indication. Similarly, the causes of ICU or readmissions could not be assessed due to the lack of detailed clinical data; however, prior studies have reported the primary causes of readmission to be AEs and disease progression [Citation34,Citation40].

In conclusion, among patients with r/r DLBCL in a large US hospital database, tisa-cel was associated with fewer inpatient admissions, shorter LOS, and more outpatient visits during the infusion encounter than axi-cel. During the entire follow-up period, rates of inpatient or ICU admission and LOS were similar between cohorts, but monthly inpatient LOS was longer for axi-cel vs. tisa-cel. Tisa-cel was associated with a higher monthly rate of outpatient visits during follow-up compared with axi-cel. Non-CAR-T costs were lower for tisa-cel than axi-cel, driven by lower inpatient costs. The rates of grade ≥3 CRS, NT, and hypogammaglobulinemia were similar between cohorts. Significantly fewer tisa-cel- vs. axi-cel-treated patients used tocilizumab. Future research with a larger patient population in a non-hospital-based database would help confirm these findings.

Author contributions

All authors contributed to the study concept, design, and data interpretation. Qing Liu, Travis Wang, Jing Zhao, and Hongbo Yang conducted data analyses. Hongbo Yang, Qing Liu, and Shelley Batts (the medical writer) wrote the manuscript. All authors critically revised the manuscript along with the medical writer and approved the current version for submission. Richard T. Maziarz is the guarantor of this manuscript.

Supplemental material

GLAL-2022-0026-File004.docx

Download MS Word (22.1 KB)

Acknowledgements

Medical writing was provided by Shelley Batts, PhD, an employee of Analysis Group, Inc. Support for this assistance was provided by Novartis Pharmaceuticals Corporation.

Ethics approval and consent to participate: As the claims data used in this study were deidentified, no Institutional Review Board oversight was required.

Disclosure statement

Hongbo Yang, Qing Liu, Travis Wang, and Jing Zhao are employees of Analysis Group, Inc., which has received consulting fees from Novartis Pharmaceuticals Corporation. Soyon Lee, Stephen Lim, Anand Dalal, and Vamsi Bollu are employees of Novartis Pharmaceuticals Corporation and hold stock/options. Richard T. Maziarz is an advisor or consultant for AlloVir, Artiva, CRISPR Therapeutics, Incyte, and Novartis; reports honoraria from Incyte, and Intellia; research support from BMS, AlloVir, and Novartis; participation in a data and safety monitoring board for Athersys, Vor Pharma, and Novartis; and a patent with Athersys.

Data availability statement

The data were provided to the authors under an agreement with Premier Healthcare Solution, Inc. Researchers can request access from Premier directly and pay the applicable fee (https://products.premierinc.com/applied-sciences/solutions).

Additional information

Funding

This work was supported by Novartis Pharmaceuticals Corporation.

References

  • National Cancer Institute. Cancer Stat Facts: NHL — diffuse large B-cell lymphoma (DLBCL); 2021; [cited 2021 Feb 24]. Available from: https://seer.cancer.gov/statfacts/html/dlbcl.html
  • Smith A, Howell D, Patmore R, et al. Incidence of haematological malignancy by sub-type: a report from the haematological malignancy research network. Br J Cancer. 2011;105(11):1684–1692.
  • Cheson BD, Nowakowski G, Salles G. Diffuse large B-cell lymphoma: new targets and novel therapies. Blood Cancer J. 2021;11(4):1–10.
  • Friedberg JW. Relapsed/refractory diffuse large B-cell lymphoma. Hematology Am Soc Hematol Educ Program. 2011;2011:498–505.
  • Crump M, Neelapu SS, Farooq U, et al. Outcomes in refractory diffuse large B-cell lymphoma: results from the International SCHOLAR-1 study. Blood. 2017;130(16):1800–1808.
  • Hamadani M, Hari PN, Zhang Y, et al. Early failure of frontline rituximab-containing chemo-immunotherapy in diffuse large B cell lymphoma does not predict futility of autologous hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2014;20(11):1729–1736.
  • Gisselbrecht C, Schmitz N, Mounier N, et al. Rituximab maintenance therapy after autologous stem-cell transplantation in patients with relapsed CD20(+) diffuse large B-cell lymphoma: final analysis of the collaborative trial in relapsed aggressive lymphoma. J Clin Oncol. 2012;30(36):4462–4469.
  • Van Den Neste E, Schmitz N, Mounier N, et al. Outcome of patients with relapsed diffuse large B-cell lymphoma who fail second-line salvage regimens in the international CORAL study. Bone Marrow Transplant. 2016;51(1):51–57.
  • Schuster SJ, Bishop MR, Tam CS, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 2019;380(1):45–56.
  • Gisselbrecht C, Glass B, Mounier N, et al. Salvage regimens with autologous transplantation for relapsed large B-cell lymphoma in the rituximab era. J Clin Oncol. 2010;28(27):4184–4190.
  • Crump M. Management of relapsed diffuse large B-cell lymphoma. Hematol Oncol Clin North Am. 2016;30(6):1195–1213.
  • Crump M, Kuruvilla J, Couban S, et al. Randomized comparison of gemcitabine, dexamethasone, and cisplatin versus dexamethasone, cytarabine, and cisplatin chemotherapy before autologous stem-cell transplantation for relapsed and refractory aggressive lymphomas: NCIC-CTG LY. 12. J Clin Oncol. 2014;32(31):3490–3496.
  • Sermer D, Batlevi C, Palomba ML, et al. Outcomes in patients with DLBCL treated with commercial CAR T cells compared with alternate therapies. Blood Adv. 2020;4(19):4669–4678.
  • Coiffier B, Sarkozy C. Diffuse large B-cell lymphoma: R-CHOP failure-what to do? Hematology Am Soc Hematol Educ Program. 2016;2016(1):366–378.
  • Chaganti S, Illidge T, Barrington S, et al. Guidelines for the management of diffuse large B-cell lymphoma. Br J Haematol. 2016;174(1):43–56.
  • Tilly H, Gomes da Silva M, Vitolo U, et al. Diffuse large B-cell lymphoma (DLBCL): ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26(Suppl. 5):v116–v125.
  • Abramson JS, Palomba ML, Gordon LI, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020;396(10254):839–852.
  • Locke FL, Ghobadi A, Jacobson CA, et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1–2 trial. Lancet Oncol. 2019;20(1):31–42.
  • 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.
  • Mian A, Wei W, Hill BT, et al. Resource utilization and factors prolonging hospitalization for patients with relapsed and refractory large B-cell lymphoma receiving tisagenlecleucel versus axicabtagene ciloleucel. Blood. 2020;136(Suppl. 1):38–39.
  • Kilgore KM, Mohammadi I, Schroeder A, et al. Medicare patients receiving chimeric antigen receptor T-cell therapy for non-Hodgkin lymphoma: a first real-world look at patient characteristics, healthcare utilization and costs. Blood. 2019;134(Suppl. 1):793.
  • Lyman GH, Nguyen A, Snyder S, et al. Economic evaluation of chimeric antigen receptor T-cell therapy by site of care among patients with relapsed or refractory large B-cell lymphoma. JAMA Netw Open. 2020;3(4):e202072.
  • Hernandez I, Prasad V, Gellad WF. Total costs of chimeric antigen receptor T-cell immunotherapy. JAMA Oncol. 2018;4(7):994–996.
  • Yang H, Hao Y, Chai X, et al. Estimation of total costs in patients with relapsed or refractory diffuse large B-cell lymphoma receiving tisagenlecleucel from a US hospital's perspective. J Med Econ. 2020;23(9):1016–1024.
  • Zhao J, Bollu V, Yang H, et al. Healthcare resource use (HRU) by infusion setting of chimeric antigen receptor T-cell (CAR-T) in patients with relapsed and refractory (r/r) diffuse large B-cell lymphoma (DLBCL): a retrospective cohort study using CMS 100% Medicare Database. J Clin Oncol. 2021;39(15 Suppl.):e19550.
  • United States Food and Drug Administration (FDA). Highlights of prescribing information: YESCARTA (axicabtagene ciloleucel); 2017; [cited 2021 Feb 24]. Available from: https://www.fda.gov/media/108377/download
  • United States Food and Drug Administration (FDA). Highlights of prescribing information: KYMRIAH (tisagenlecleucel); 2018; [cited 2019 Oct 28]. Available from: https://www.fda.gov/files/vaccines%2C%20blood%20%26%20biologics/published/Package-Insert---KYMRIAH.pdf
  • Premier Applied Sciences. Premier Healthcare Database: data that informs and performs [white paper]; 2020; [cited 2021 Sep 1]. Available from: https://products.premierinc.com/downloads/PremierHealthcareDatabaseWhitepaper.pdf
  • Riedell PA, Walling C, Nastoupil LJ, et al. A multicenter retrospective analysis of outcomes and toxicities with commercial axicabtagene ciloleucel and tisagenlecleucel for relapsed/refractory aggressive B-cell lymphomas. Biol Blood Marrow Transplant. 2020;26(3):S41–S42.
  • Pasquini MC, Hu ZH, Curran K, et al. Real-world evidence of tisagenlecleucel for pediatric acute lymphoblastic leukemia and non-Hodgkin lymphoma. Blood Adv. 2020;4(21):5414–5424.
  • Pasquini MC, Locke FL, Herrera AF, et al. Post-marketing use outcomes of an anti-CD19 chimeric antigen receptor (CAR) T cell therapy, axicabtagene ciloleucel (axi-cel), for the treatment of large B cell lymphoma (LBCL) in the United States (US). Blood. 2019;134(Suppl. 1):764.
  • Jacobson CA, Locke FL, Hu Z-H, et al. Real-world evidence of axicabtagene ciloleucel (axi-cel) for the treatment of large B-cell lymphoma (LBCL) in the United States (US). J Clin Oncol. 2021;39(15 Suppl.):7552.
  • Klink A, Savill K, Liassou D, et al. Real-world treatment with CAR T-cell therapy of United States (US) patients with large B cell lymphoma (LBCL). European Hematology Association Congress (virtual); 2021 Jun 9–19.
  • Myers GD, Verneris MR, Goy A, et al. Perspectives on outpatient administration of CAR-T cell therapy in aggressive B-cell lymphoma and acute lymphoblastic leukemia. J Immunother Cancer. 2021;9(4):e002056.
  • Harris A, Hohmann S, Epting G, et al. Quality and cost outcomes in chimeric antigen receptor T-cell immunotherapy in adult large B-cell cancer patients from the Vizient Clinical Database. Biol Blood Marrow Transplant. 2020;26(3):S316–S317.
  • Harris A, Hohmann S, Dolan C. Real-world quality and cost burden of cytokine release syndrome requiring tocilizumab or steroids during CAR-T infusion encounter. Biol Blood Marrow Transplant. 2020;26(3):S312.
  • Maziarz R, Appel P, Cota B, et al. Abstract #370: financial impact of effector cell therapy on the care of patients with advanced B-cell malignancies. 2021 Transplantation & Cellular Therapy Meeting (virtual); 2021 Feb 8–12.
  • Jaglowski S, Hu Z-H, Zhang Y, et al. Tisagenlecleucel chimeric antigen receptor (CAR) T-cell therapy for adults with diffuse large B-cell lymphoma (DLBCL): real world experience from the Center for International Blood & Marrow Transplant Research (CIBMTR) cellular therapy (CT) registry. Blood. 2019;134(Suppl. 1):766.
  • Riedell PA, Brower J, Nastoupil L, et al. A multicenter analysis of outcomes, toxicities, and patterns of use with commercial axicabtagene ciloleucel and tisagenlecleucel for relapsed/refractory aggressive B-cell lymphomas. ASH Annual Meeting; 2021 Dec 11–14; Atlanta, GA; 2021.
  • Sharma A, Singh V, Deol A. Epidemiology and predictors of readmission in CAR T-cell therapy recipients. Blood. 2021;138(Suppl. 1):3010.