Skip to main content
Sage Choice logoLink to Sage Choice
. 2024 Mar 14;44(3):296–306. doi: 10.1177/0272989X241234070

CAR T-cell Therapy for Diffuse Large B-cell Lymphoma in Canada: A Cost-Utility Analysis

Lisa Masucci 1,*,, Feng Tian 2,*, Stephen Tully 3, Zeny Feng 4, Tom McFarlane 5, Kelvin K W Chan 6,7,*, William W L Wong 8,9,*
PMCID: PMC10988988  PMID: 38486447

Abstract

Background

Chimeric antigen receptor (CAR) T-cell therapy is a novel cell therapy for treating non-Hodgkin lymphoma. The development of CAR T-cell therapy has transformed oncology treatment by offering a potential cure. However, due to the high cost of these therapies, and the large number of eligible patients, decision makers are faced with difficult funding decisions. Our objective was to assess the cost-effectiveness of tisagenlecleucel for adults with relapsed/refractory diffuse large B-cell lymphoma in Canada using updated survival data from the recent JULIET trial.

Methods

We developed an individual-simulated discrete event simulation model to assess the costs and quality-adjusted life-years (QALY) of tisagenlecleucel compared with salvage chemotherapy. Survival estimates were obtained from a published clinical trial and retrospective analysis. If patients remained progression free for 5 y, they were assumed to be in long-term remission. Costing and utility data were obtained from reports and published sources. A Canadian health care payer perspective was used, and outcomes were modeled over a lifetime horizon. Costs and outcomes were discounted at 1.5% annually, with costs reported in 2021 Canadian dollars. A probabilistic analysis was used, and model parameters were varied in 1-way sensitivity analyses and scenario analyses.

Results

After we incorporated the latest clinical evidence, tisagenlecleucel led to an additional cost of $503,417 and additional effectiveness of 2.48 QALYs, with an incremental cost-effectiveness ratio of $202,991 compared with salvage chemotherapy. At a willingness-to-pay threshold of $100,000/QALY, tisagenlecleucel had a 0% likelihood of being cost-effective.

Conclusions

At the current drug price, tisagenlecleucel was not found to be a cost-effective option. These results heavily depend on assumptions regarding long-term survival and the price of CAR T. Real-world evidence is needed to reduce uncertainty.

Highlights

  • For patients with diffuse large B-cell lymphoma who failed 2 or more lines of systemic therapy, CAR T was not found to be a cost-effective treatment option at a willingness-to-pay threshold of $100,000.

  • These results heavily depend on the expected long-term survival. The uncertainty in the model may be improved using real-world evidence reported in the future.

Keywords: chimeric antigen receptor T-cell therapy, tisagenlecleucel, cost-effectiveness analysis


In 2019, it was estimated that there were 10,000 new cases of non-Hodgkin lymphoma (NHL) in Canada.1,2 Diffuse large B-cell lymphoma (DLBCL) is the most common subtype, accounting for 30% to 40% of newly diagnosed lymphoma cases.1,2 Of these patients, 60% will receive first line chemoimmunotherapy and be successfully treated.35 The remaining will experience relapsed or refractory (r/r) disease and require subsequent therapy. Standard therapy for r/r DLBCL is salvage chemotherapy followed by high-dose therapy and autologous stem-cell transplantation (SCT). 6 Following this treatment, only 30% will achieve long-term remission, and more than 50% will relapse. 7 Few treatment options remain after autologous SCT, and treatment often results in poor outcomes. 8 Chimeric antigen receptor (CAR) T-cell therapy has emerged as an innovative and promising therapy in the management of r/r DLBCL. CAR T-cell therapy is a personalized therapy in which a patient’s own T-cells are harvested, modified to specifically target cancer cells, and readministered to the patient, where they bind to and kill cancer cells.9,10

Tisagenlecleucel (tisa-cel) is a type of CAR T-cell therapy approved for use in select adult lymphoma populations. The treatment targets a CD-19 antigen that is expressed in more than 95% of B-cell malignancies. 7 Tisa-cel was approved by the United States Food and Drug Administration in May 2018 and by Health Canada in May 2019.11,12 The indication for this CAR T-cell product is adult patients with r/r DLBCL after 2 or more lines of systemic therapy.11,12 In 2019, Canada’s national health technology assessment agency, the Canadian Agency for Drugs and Technologies in Health (CADTH), recommended funding tisa-cel conditional on a substantial price reduction. 13 This decision was based on an analysis that used short-term survival data with a median follow-up of 14 mo.14,15

In an updated survival analysis, at a median follow-up of 40.3 mo, tisa-cel resulted in an overall response rate of 53% and complete response of 45%. 16 While the single-arm study showed improved survival in select patients, 16 the high cost of CAR T-cell therapy ($513,655 per patient), 17 together with the substantial use of health care resources, make this therapy a challenging drug reimbursement problem. 18 The cost-effectiveness of tisa-cel, using the updated survival analysis figures, is currently unknown. The objective of this study was to use the updated survival estimates to evaluate the cost-utility of tisa-cel in adult patients with r/r DLBCL after 2 or more lines of systemic therapy.

Methods

Model Overview

We developed an individual-simulated discrete event simulation (DES) model using R (version 4.2.1) to assess the long-term costs and outcomes of patients with r/r DLBCL who received CAR T-cell therapy compared with a similar population who received a salvage chemotherapy regimen (rituximab, gemcitabine, dexamethasone, and cisplatin [R-GDP]). DES models can simulate individual patients as they transit through a health care system, with transitions dependent on both patient characteristics and the availability of resources. These types of models can determine if queues will develop due to resource constraints and can also evaluate the impact of wait-time strategies. 19 In Canada, wait times are on average 1 mo after apheresis, as cells need to be shipped to a manufacturer in the United States. The DES model allows us to capture the wait times that patients may experience. A schematic of the treatment pathway and structure of the model is shown in Supplementary Figure 1. The model simulates individual patients, their health states related to treatment, and clinical states to reflect the natural history of DLBCL. A simulated cohort of 2,000 patients was followed starting at a mean age of 58 y. The start ages were randomly sampled based on a normal distribution with a mean of 58 y for all simulated patients. This start age aligns with the median age reported in the clinical trial. 20 The model consists of 3 main health states: 1) progression free, 2) progressed, and 3) long-term remission. A patient enters the model in the progression-free health state and can remain progression free or move to the progressed, long-term remission or death health states over time. A person can only enter the long-term remission health state after 5 y of being progression free. 21 Individuals who progress after treatment have the opportunity to receive autologous SCT after salvage chemotherapy and allogeneic SCT after CAR T-cell therapy.14,22

Events included in the model are grade 3/4 adverse events reported in the clinical trials (e.g., cytokine release syndrome, neurologic events) and death. Wait times were also modeled using a fixed queue (first in, first out) in our simulation and represented the amount of time that a patient must wait to receive CAR T-cell therapy (base case = 1 mo). This was to reflect delays, such as extra manufacturing time, traveling and delivery time, waiting for specialists, and reimbursement approval by public health care payers.

All patients in the model eventually died from DLBCL or background mortality. A lifetime time horizon was taken, and future costs and benefits were discounted at 1.5% annually based on published Canadian guidelines. 23 An economic analysis plan was developed for this study and is available online through the Canadian Institutes of Health Research.

Treatment Strategies

We considered 2 treatment strategies: 1) salvage chemotherapy (R-GDP) and 2) CAR T-cell therapy (tisa-cel). There are several other chemotherapy combinations for salvage chemotherapy available. These include rituximab, dexamethasone, cytarabine, and cisplatin (R-DHAP) and less commonly rituximab, ifosfamide, carboplatin, and etoposide (R-ICE) 3 ; however, the most common treatment in the Canadian setting for adult patients with DLBCL is R-GDP. 24 Therefore, we assumed that R-GDP was used for all patients in the salvage chemotherapy arm.

Model Inputs

Survival data

Overall survival (OS) and progression-free survival (PFS) data were collected from the published findings of trials investigating CAR T-cell therapies for patients with r/r DLBCL. The JULIET trial was a multicenter, single-arm, phase 2 study that used tisa-cel to treat patients with r/r DLBCL and followed patients up to a median of 14 mo. 15 A long-term follow-up analysis was published with a median follow-up of 40.3 mo. 16 For the salvage chemotherapy arm, data from the SCHOLAR-1 trial were used for OS. SCHOLAR-1 is a retrospective study that consisted of 636 relevant patients undergoing mixed salvage chemotherapy regimens across 4 cohorts. A subset of patients who had a similar Eastern Cooperative Oncology Group performance status (score 0–1) to patients in 2 CAR T-cell therapy trials (ZUMA-1 and JULIET) were examined. 8 Since SCHOLAR-1 reported only OS, we used a proportional relationship based on published PFS and OS curves for patients using rituximab, dexamethasone, cytarabine, and cisplatin (R-DHAP) to derive the PFS curve.2527

To extrapolate survival data beyond the trial period, the published OS and PFS curves from the clinical trials were digitized. An algorithm developed by Guyot et al. 28 was applied to reconstruct the individual patient data underlying the OS and PFS curves. A number of parametric functions were fitted onto the OS and PFS curves. Standard parametric models (exponential, Weibull, lognormal, loglogistic, and gamma distributions) were fitted to the data, and the one with the lowest Bayesian information criterion was chosen. Since the plateau of the survival curve is difficult to accommodate with a single parametric function, we used piecewise functions that connected 2 parametric functions based on overall goodness of fit. 29 This approach allowed us to capture the structural changes of the survival curves. Using the best-fitted parametric models, the time to disease progression and death were sampled from the quantile function for each patient. 19 Supplementary Table 1 lists the details of the distributions used for the survival curves needed by the model to compute the monthly transition probabilities between the disease progression and survival health states.

Patients who survived beyond 5 y were considered to be in long-term remission. Patients in this health state could die of background mortality. This assumption was based on 2 studies that assessed long-term outcomes in patients with DLBCL and the low probability of progression past this point.21,26 The survival estimates of the general population were derived from the 2017 to 2019 Statistics Canada Life Table. 30

Adverse events

The administration of CAR T-cells is associated with significant short-term and long-term side effects. During the first month, common adverse events include cytokine release syndrome (CRS), cytopenia, infection, and febrile neutropenia. Grade 3 or higher CRS is seen in up to 23% of patients who received tisa-cel. 16 Neurologic events (i.e., encephalopathy), grade 3 or higher have been reported in up to 4% with tisa-cel. 16 Grade 3 or higher CRS commonly requires an intensive care unit (ICU) admission. Thus, patients must be closely monitored for severe side effects such as CRS after infusion. In addition to these adverse events, we considered other grade 3/4 events reported for tisa-cel if they affected more than 10% of patients in the trial. Although less than 10% of patients who received CAR T-cell therapy experienced grade 3/4 nausea, fatigue, and vomiting, we added these adverse events to the model to keep the reporting consistent with the salvage chemotherapy arm.

For salvage chemotherapy, adverse events were not reported in the clinical trial used for the efficacy data. It was assumed that the adverse events that occurred with this treatment matched a clinical trial in which patients with r/r B-cell lymphoma were randomized to receive GDP or DHAP. 31 Table 1 lists the adverse events considered in our model along with their probabilities.

Table 1.

Monthly Probabilities Related to Treatment and Adverse Events

Parameter Value Range Distribution Reference
Proportion of patients receiving bridging chemotherapy
 Tisa-cel 0.90 0.68–1.00 Beta Schuster et al. 16
Proportion of patients receiving SCT
 Tisa-cel 0.05 0.04–0.06 Beta CADTH 14
 Salvage chemotherapy 0.30 0.22–0.37 Beta Corbett et al. 22
Adverse events (grade 3–4) with tisa-cel
 Cytokine release syndrome 0.23 0.17–0.28 Beta Schuster et al. 16
 B-cell aplasia 0.30 0.23–0.38 Beta Schuster et al. 16
 Febrile neutropenia 0.17 0.12–0.21 Beta Schuster et al. 16
 Anemia 0.39 0.29–0.49 Beta Schuster et al. 16
 Encephalopathy 0.04 0.03–0.05 Beta Schuster et al. 16
 Hypotension 0.09 0.07–0.11 Beta Schuster et al. 16
 Neutropenia 0.20 0.15–0.25 Beta Schuster et al. 16
 Pyrexia 0.05 0.04–0.07 Beta Schuster et al. 16
 Thrombocytopenia 0.12 0.09–0.15 Beta Schuster et al. 16
 White blood cell count decreased 0.32 0.24–0.40 Beta Schuster et al. 16
 Infections 0.19 0.14–0.24 Beta Schuster et al. 16
 Nausea 0.01 0.01–0.01 Beta Schuster et al. 16
 Fatigue 0.06 0.05–0.08 Beta Schuster et al. 16
Adverse events with salvage chemotherapy
 Febrile neutropenia 0.23 0.17–0.29 Beta Crump et al. 31
 Infections 0.09 0.07–0.11 Beta Crump et al. 31
 Nausea 0.08 0.06–0.10 Beta Crump et al. 31
 Vomiting 0.07 0.05–0.09 Beta Crump et al. 31
 Fatigue 0.09 0.07–0.11 Beta Crump et al. 31
Wait time for CAR T 1 mo 3–6 mo Clinician input 37

CADTH, Canadian Agency for Drugs and Technologies in Health; CAR, chimeric antigen receptor; SCT, stem-cell transplantation; tisa-cel, tisagenlecleucel.

Costs

This analysis adopted a Canadian health care payer perspective, and as such, direct health care costs were considered (Table 2). These costs included pretreatment costs, cost of treatment, monitoring, and adverse events. The pretreatment costs for CAR T-cell therapy included leukapheresis and lymphodepleting chemotherapy with fludarabine and cyclophosphamide. A proportion of patients also incurred the cost of bridging chemotherapy before receiving CAR T-cell infusion. The primary sources of data for costs included the Ontario Case Costing Initiative, the Canadian Institute for Health Information, the Ontario Schedule of Benefits and Fees for Physician Services, and the Ontario Drug Benefit.3235

Table 2.

Costs and Utilities

Parameter Value Range ($) Distribution Reference
Pretreatment
 Leukapheresis 1,441 1,081–1,801 Gamma OCCI 30
 Pretreatment tisa-cel 152 114–190 Gamma Ellis 24
 Bridging chemotherapy 20,625 15,469–25,781 Gamma CADTH 14
Treatment
 Tisa-cel 513,655 385,241–642,068 Gamma RedBook 17
 Administration of CAR T 105 79–131 Gamma SoB 31
 Hospitalization (regular ward) 1,950 1,462–2,437 Gamma CIHI 33
 Salvage chemotherapy (R-GDP, 6 cycles) 22,344 16,758–27,936 Gamma ODB 32
 Administration of chemotherapy 75 56–94 Gamma SoB 31
 Stem cell transplant 170,201 127,651–212,751 Gamma Ontario Health 35
Posttreatment follow-up and monitoring
 Office consultation, hematology 169 127–211 Gamma SoB 31
 PET scan 1,706 1,279–2,132 Gamma Ellis 24
Adverse events
 Febrile neutropenia 7,137 5,352–8,921 Gamma OCCI 30
 Infections 470 352–587 Gamma OCCI 30
 Pyrexia 447 335–558 Gamma OCCI 30
 Neutropenia 534 400–667 Gamma OCCI 30
 Anemia 8,584 6,438–10,730 Gamma OCCI 30
 Thrombocytopenia 459 344–574 Gamma OCCI 30
 Hypotension 590 442–737 Gamma OCCI 30
 White blood cell count decreased 456 342–570 Gamma OCCI 30
 Encephalopathy 4,271 3,203–5,338 Gamma OCCI 30
 Nausea 266 200–333 Gamma Ellis 24
 Vomiting 342 257–428 Gamma Ellis 24
 Fatigue 334 251–418 Gamma Ellis 24
 CRS hospitalization (intensive care unit) 3,958 2,968–4,947 Gamma CIHI 33
 Tocilizumab (2 doses) 2,553 1,915–3,191 Gamma ODB, 32 Ellis 24
 IVIG 2,556 1,917–3,195 Gamma ODB, 32 Ellis 24
Predeath costs
 Care in the last year before lymphoma-related death 58,709 58,687–58,731 Gamma De Oliveria et al., 39 Ellis 24
 Health care costs in the last year of life 60,147 45,111–75,184 Gamma Tanuseputo et al., 40 Ellis 24
Utilities
 Progression-free health state 0.80 0.53–1.00 Beta Lin et al. 43
 Progressed health state 0.72 0.39–1.00 Beta Lin et al. 42
 Disutility with tisa-cel 0.05 0–0.13 Beta Lin et al. 42
 Disutility of salvage chemotherapy 0.15 0.11–0.19 Beta CADTH 14
 Disutility of stem cell transplant 0.57 0.31–0.87 Beta Sung et al. 45
 Additional utility starting from 18 mo after treatment 0.06 0.05–0.07 Beta Patrick et al. 46

CADTH, Canadian Agency for Drugs and Technologies in Health; CAR, chimeric antigen receptor; CIHI, Canadian Institute for Health Information; CRS, cytokine release syndrome; IVIG, intravenous immunoglobulin; OCCI, Ontario Case Costing Initiative; ODB, Ontario Drug Benefit; PET, positron emission tomography; SoB, schedule of benefits; tisa-cel, tisagenlecleucel.

The cost of CAR T-cell therapy included the cost of the CAR T-cell product, physician costs for monitoring during the administration of CAR T-cell therapy, and an inpatient hospital stay. The cost of CAR T-cell therapy was based on the wholesale acquisition cost of $513,655 CAD for tisa-cel. 32 The cost of salvage chemotherapy (gemcitabine-dexamethasone-cisplatin-rituximab) was based on 6 cycles within 21 d. 24 For the cost of drugs in which the dose is weight based, an average body surface index of 1.7 m2 was used. 24 For patients who received an SCT, the cost of an adult allogeneic matched donor was applied. 36

Posttreatment monitoring included the cost of physician consultations (hematology) twice a week in the first month after infusion, weekly in the second month, monthly thereafter up to 6 mo, every 3 mo for up to 2 y, and every 6 mo until 5 y.37,38 The cost of a positron emission tomography scan was applied at 3 mo, 6 mo, 1 y, and yearly up to 5 y. 24

We included the cost of the top grade 3 or 4 adverse events reported in each CAR T-cell therapy clinical study. The most significant adverse events associated with CAR T-cell therapy are CRS and B-cell aplasia. The cost of CRS was calculated by multiplying the cost of an ICU stay and 2 doses of tocilizumab by the time to CRS resolution reported in the clinical trials. The average cost of an ICU stay was obtained from the Canadian Institute for Health Information. 35 For those who experienced B-cell aplasia, the cost of intravenous immune globulin was applied monthly up until 1 y. 13 The costs of all other adverse events were based on hospital costs obtained from the Ontario Case Costing Initiative, which reports the costs of inpatient hospital visits in Ontario. 32

For all patients who died, the predeath health care costs in the last year before the lymphoma-related death were applied.39,40 These predeath costs were obtained from 2 retrospective cohort studies and included any health care visits, hospitalizations, medications, and home care visits paid for by Ontario Health. All costs were inflated to 2021 Canadian dollars using the Consumer Price Index for health care services in Ontario. 41 The United States prices of CAR T-cell therapy were converted to Canadian dollars using the purchasing power parity exchange rate. 42

Utilities

Utility values (Table 2) for the progression-free and progressed health states were obtained from published quality-of-life data, based on a safety management cohort from the ZUMA-1 trial, which were generated using the EQ-5D-5L instrument. 43 The ZUMA-1 trial focused on patients treated with axicabtagene ciloleucel (axi-cel), which is a similar product to tisa-cel. A total of 34 patients with r/r DLBCL treated with axi-cel were initially part of the quality-of-life study, with a median follow-up of 5.1 mo. The mean EQ-5D-5L index was 0.80 (SD = 0.14) for the progression-free health state and 0.72 (SD = 0.17) for progressed disease. 43 To capture short-term adverse events associated with treatment, we applied a disutility while patients were receiving treatment. For CAR T-cell therapy, the disutility associated with treatment was 0.05 (SE = 0.04) and applied over a 1-y period at the time of the infusion. 43 For those who experienced grade 3/4 CRS, health utility was reduced to 0 for the first 2 mo after the CRS-related event. 43 For salvage chemotherapy, a disutility of 0.15 was applied for the treatment (6 cycles of chemotherapy),14,44 based on a randomized multicenter study comparing CHOP with CHOP plus G-CSF.14,44 The disutility associated with SCT (0.57) was obtained from a study examining the quality of life of young adults with acute myeloid leukemia who had an allogeneic bone marrow transplantation. 45 This disutility was applied at the time of therapy. An additional utility of 0.06 starting 18 mo after treatment was applied, as a study examining the quality of life of patients who received CAR T-cell therapy indicated that the utilities increase at this time point. 46

Analysis

The DES model captured patient-level variability and parameter-level uncertainty. For the base case, a probabilistic sensitivity analysis was conducted to account for the uncertainty of all the parameters. Parameters were sampled from appropriate distributions (e.g., beta distribution for probabilities) informed by the corresponding ranges. The ranges were based on the published 95% confidence intervals or, in the absence of literature, ±25% from their base case value. We simulated 2,000 individuals and performed 1,000 simulations for each treatment strategy to estimate the mean outcomes. We estimated the likelihood of each strategy being more favorable across a range of willingness-to-pay (WTP) thresholds using cost-effectiveness acceptability curves (CEACs). An upper limit of $100,000 per QALY was used as a cost-effectiveness threshold, as this threshold is commonly used by Canada’s national health technology assessment agency. 14

Deterministic sensitivity analysis

To assess the impact of each parameter on the results, we conducted deterministic 1-way sensitivity analyses, varying the input parameters one at a time according to their 95% confidence intervals, or ±25% from their base case value.

Scenario analysis

Scenario analyses were conducted on the time to achieve long-term remission, mortality in the long-term remission health state (standardized mortality ratio of 1.09), 47 alternative parametric distribution (i.e., second best fit) for the OS and PFS extrapolation, and alternative wait time to receive CAR T-cell therapy. We also conducted a scenario analysis using survival and cost estimates for axi-cel, which is another CAR T-cell therapy approved in Canada.20,48,49 The survival curves for axi-cel can be found in Supplementary Figure 4 and the parameter estimates in Supplementary Table 2.

Validation

To validate our model, the Kaplan-Meier survival curves for the OS and PFS from the trials were plotted alongside our model outcomes. We visually inspected the overlay by comparing the OS and PFS curves from the model and the original trials. Supplementary Figures 2 and 3 demonstrate a comparison between the trial data and the output of our simulation when comparing the JULIET and SCHOLAR-1 trials for OS. The face validity of the model structure, assumptions, data, and results were validated with clinical experts.

Results

Base Case Analysis

The results of the base case analysis are presented in Table 3. In the probabilistic analysis, tisa-cel compared with salvage chemotherapy resulted in an additional cost of $503,417 and additional effectiveness of 2.48 QALYs. This led to an incremental cost-effectiveness ratio (ICER) of $202,991 per QALY gained. Tisa-cel was not cost-effective based on a WTP threshold of $100,000 per QALY. Our model estimated 14.8% of cancer-related deaths were averted due to the treatment at 5 y. The results demonstrated that for tisa-cel, 100% of the 5,000 second-order simulated ICERs were in the northwest quadrant, meaning that tisa-cel was more costly and more effective compared with salvage chemotherapy (Figure 1). The CEAC demonstrated that compared with salvage chemotherapy, tisa-cel had a 0% probability of being cost-effective at $100,000/QALY (Figure 2).

Table 3.

Base Case Analysis Results

Salvage Chemotherapy Tisa-cel
Total expected cost, 2021 CAD $143,115 $637,532
Incremental cost $503,417
Total expected QALYs 3.55 6.03
Incremental QALYs 2.48
Total expected life-years 5.64 9.15
Incremental life-years 3.51
ICER (cost per QALY) $202,991

Figure 1.

Figure 1

Cost-effectiveness plane for tisagenlecleucel (tisa-cel) versus salvage chemotherapy.

Figure 2.

Figure 2

Cost-effectiveness acceptability curve for tisagenlecleucel (tisa-cel) versus salvage chemotherapy.

Deterministic Sensitivity Analysis

Figure 3 demonstrates the top 10 variables that affected the ICERs. The ICER ranged from $152,632 per QALY gained to $444,535 per QALY gained. The model was most sensitive to the time horizon, the starting age of the cohort, discount rate, cost of the product, and the utility of the progression-free health state. Tisa-cel was not cost-effective in all of the one-way sensitivity analyses; however, it came closer to the cost-effectiveness threshold when the cost of the product was reduced to $385,241 and the starting age of the cohort was reduced to 43.5 y. In our analysis, the price of tisa-cel would need to be reduced by 51% ($251,602) to meet the $100,000 WTP threshold.

Figure 3.

Figure 3

Deterministic sensitivity analysis results.

Scenario Analysis

The results of the scenario analysis are presented in Supplementary Table 3. A change in the time required to achieve long-term remission, increasing the mortality for those in long-term remission, and the wait time to receive CAR T-cell therapy did not affect our assessment of the economic value of tisa-cel. The results were also robust to changes in the OS and PFS curves (i.e., using the second-best fit). Using the survival and cost estimates associated with axi-cel resulted in an ICER of $105,649/QALY compared with salvage chemotherapy, which is closer to our WTP threshold. If the cost of CAR T-cell therapy was reduced to $251,602, tisa-cel would be cost-effective compared with salvage chemotherapy at a WTP threshold of $100,000/QALY.

Discussion

CAR T-cell therapy has the potential to improve patient lives by inducing a long-term durable remission in patients who have run out of treatment options; however, this therapy comes at a high cost. After using longer-term survival data, our results suggest that tisa-cel would not be cost-effective using a WTP threshold of $100,000 per QALY. The ICERs were driven by the time horizon, starting age of the cohort, discount rate, price of the product, and assumptions regarding long-term survival.

Our results are consistent with previous economic studies.14,50,51 Three studies found tisa-cel to be cost-effective compared with salvage chemotherapy; however, there are key differences between these studies and ours.5254 Two of these studies relied on less mature data from the JULIET trial and selected a 3-year cure point.53,54 In our scenario analysis, if we reduced the cure point from 5 to 3 y, the ICER of tisa-cel versus salvage chemotherapy would be reduced to $169,083/QALY, bringing the ICER closer to the WTP threshold. A study from the United States that relied on more mature data from the JULIET trial and used a 5-y cure rate found tisa-cel to be cost-effective. 52 However, there are a few key differences between our study and theirs, which likely had a cumulative impact on the ICER. First, Qi et al. reported the total costs associated with subsequent SCT to be $105,000 USD more with the salvage chemotherapy arm compared with the tisa-cel arm. In our model, subsequent SCT was associated with $42,000 CAD more in costs. 52 Our cost of SCT and rates of subsequent transplant were lower as we considered only autologous SCT for the salvage chemotherapy arm and allogeneic SCT for the tisa-cel arm. This is in line with costs and clinical practice in Canada. To be eligible for CAR T in Canada, the patient would not have previously received allogeneic SCT; however, this treatment may be offered after CAR T. As our tornado diagram demonstrates, if we increased our rate of subsequent SCT for salvage chemotherapy to 37.3%, our ICER would be $193,072 per QALY gained, which is much lower than our base case. Second, the total cost after progression for the salvage chemotherapy therapy arm and the total cost of terminal care were larger for the salvage chemotherapy arm in the study by Qi et al. 52 This is a function of more patients progressing and requiring SCT in the salvage chemotherapy arm. Finally, Qi et al. 52 reported greater incremental QALYs gained with tisa-cel versus our study, which is a result of the disutility associated with subsequent SCT and progression. All of these factors together resulted in an ICER that is more favorable than ours.

To our knowledge, this is the first study to use a DES model to estimate the cost-effectiveness of tisa-cel compared with salvage chemotherapy. This type of model further enhanced the realism of the simulations by allowing costs and effectiveness to accumulate and be discounted accurately over a continuous period of time. Another key strength of our analysis is including wait times for treatment in the model, which have been shown to affect the ICER. Our study is further enhanced by the use of more recent survival data from the JULIET trial. 16 Finally, we used a 2-phase parametric curve fitting, which greatly improved the model fit and the accuracy of our extrapolated estimates. This DES model will be used in the future to evaluate the cost-effectiveness of tisa-cel using real-world evidence so that results are more generalizable to public payers. This DES model will allow us to account for the impact of time delays to receiving treatment in the real world (e.g., survival, additional resources used).

Limitations

There are some limitations to this cost-effectiveness analysis. First, since no head-to-head trial data were available, this analysis relied on data sources from a single-arm trial. Data from a real-world study could help to inform the true estimates. Second, data from this single-arm trial included a median of 40.3 mo of follow-up, and so assumptions had to be made regarding the extrapolation of outcomes beyond this point and the percentage of patients who are deemed cured. Longer-term data can help to inform more precise estimates of cost-effectiveness over a patient’s lifetime. The collection of standardized outcomes in a registry is needed to better assess the long-term effectiveness, safety, and cost-effectiveness. Third, similar to other published cost-effectiveness analyses, we calibrated our DES model using published OS and PFS estimates. We did not have access to the individual patient-level data, which limits our ability to accurately estimate the exact timing of each event individually, which can cause us to overestimate or underestimate our results. A series of scenario analyses were run using the second best fit for the OS and PFS curves. Fourth, we assumed that after 5 y of being progression free, a patient was cured. Longer follow-up data will be needed to inform this estimate. Lastly, the utility estimates informing our study were mainly based on the quality of life of patients who received axi-cel. Although this treatment is similar to tisa-cel, future studies are necessary to validate these estimates.

Conclusion

Our study suggests that, compared to salvage chemotherapy, tisa-cel was not a cost-effective treatment option for patients with DLBCL who failed 2 or more lines of systemic therapy at a WTP threshold of $100,000/QALY. Our analysis indicated that there is still a reasonable amount of uncertainty in terms of survival and utility estimates. Given that CAR T-cell therapy has already been introduced in many jurisdictions around the world, further research based on real-world evidence can address some of the uncertainty raised in our analysis.

Supplemental Material

sj-docx-1-mdm-10.1177_0272989X241234070 – Supplemental material for CAR T-cell Therapy for Diffuse Large B-cell Lymphoma in Canada: A Cost-Utility Analysis

Supplemental material, sj-docx-1-mdm-10.1177_0272989X241234070 for CAR T-cell Therapy for Diffuse Large B-cell Lymphoma in Canada: A Cost-Utility Analysis by Lisa Masucci, Feng Tian, Stephen Tully, Zeny Feng, Tom McFarlane, Kelvin K. W. Chan and William W. L. Wong in Medical Decision Making

Footnotes

Author Contributions: Concept and design: KC, WW, TF, ZF

Acquisition of data: LM, FT, ST

Statistical analysis: LM, FT, ST

Interpretation of data: All

Drafting of the manuscript: LM, FT, ST

Critical revision of the paper: All

Supervision: KC, WW

Approval of submission: All

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article. The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: Financial support for this study was provided by the Canadian Institutes of Health Research PJK-175385 and PJK-179825 and as well as an award provided by Medicine by Design through the Canada First Research Excellence Fund. The funding agreement ensured the author’s independence in designing the study, interpreting the data, writing, and publishing the report.

Contributor Information

Lisa Masucci, Toronto Health Economics and Technology Assessment Collaborative, Toronto General Hospital, ON, Canada.

Feng Tian, School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.

Stephen Tully, School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.

Zeny Feng, Department of Mathematics and Statistics, University of Guelph, Guelph, ON, Canada.

Tom McFarlane, School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.

Kelvin K. W. Chan, Sunnybrook Odette Cancer Centre, Toronto, ON, Canada; Canadian Centre for Applied Research in Cancer Control, Toronto, ON, Canada.

William W. L. Wong, Toronto Health Economics and Technology Assessment Collaborative, Toronto General Hospital, ON, Canada; School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.

References

  • 1. Canadian Cancer Society. Diffuse large B-cell lymphoma. 2019. [Available from: Accessed 20 October, 2022]. [Google Scholar]
  • 2. Levine BL, Miskin J, Wonnacott K, Keir C. Global manufacturing of CAR T cell therapy. Mol Ther Methods Clin Dev. 2017;4:92–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Gisselbrecht C, Van Den Neste E. How I manage patients with relapsed/refractory diffuse large B cell lymphoma. Br J Haematol. 2018;182(5):633–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Sehn LH, Gascoyne RD. Diffuse large B-cell lymphoma: optimizing outcome in the context of clinical and biologic heterogeneity. Blood. 2015;125(1):22–32. [DOI] [PubMed] [Google Scholar]
  • 5. Staton AD, Koff JL, Chen Q, Ayer T, Flowers CR. Next-generation prognostic assessment for diffuse large B-cell lymphoma. Future Oncol. 2015;11(17):2443–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Skrabek P, Assouline S, Christofides A, et al. Emerging therapies for the treatment of relapsed or refractory diffuse large B cell lymphoma. Curr Oncol. 2019;26(4):253–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Jain MD, Bachmeier CA, Phuoc VH, Chavez JC. Axicabtagene ciloleucel (KTE-C19), an anti-CD19 CAR T therapy for the treatment of relapsed/refractory aggressive B-cell non-Hodgkin’s lymphoma. Ther Clin Risk Manag. 2018;14:1007–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. 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):1800–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Benmebarek MR, Karches CH, Cadilha BL, Lesch S, Endres S, Kobold S. Killing mechanisms of chimeric antigen receptor (CAR) T cells. Int J Mol Sci. 2019;20(6):1283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Daniyan AF, Brentjens RJ. At the bench: chimeric antigen receptor (CAR) T cell therapy for the treatment of B cell malignancies. J Leukoc Biol. 2016;100(6):1255–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Government of Canada. Regulatory Decision Summary—Kymriah. Ottawa (Canada): Government of Canada; 2019. [Google Scholar]
  • 12. US Food and Drug Administration. Kymriah (tisagenlecleucel). Silver Spring (MD): US Food and Drug Adminsitration; 2019. [Google Scholar]
  • 13. Canadian Agency for Drugs and Technologies in Health. Tisagenlecleucel for Acute Lymphoblastic Leukemia and Diffuse Large B-cell Lymphoma: Recommendations. Ottawa (Canada): Canadian Agency for Drugs and Technologies in Health; 2019. [Google Scholar]
  • 14. Canadian Agency for Drugs and Technologies in Health. Tisagenlecleucel for Diffuse Large B-cell Lymphoma: Economic Review Report. Ottawa (Canada): Canadian Agency for Drugs and Technologies in Health; 2019. [PubMed] [Google Scholar]
  • 15. 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. [DOI] [PubMed] [Google Scholar]
  • 16. Schuster SJ, Tam CS, Borchmann P, et al. Long-term clinical outcomes of tisagenlecleucel in patients with relapsed or refractory aggressive B-cell lymphomas (JULIET): a multicentre, open-label, single-arm, phase 2 study. Lancet Oncol. 2021;22(10):1403–15. [DOI] [PubMed] [Google Scholar]
  • 17. Merative. IBM Micromedex RED BOOK Online. 2022. Available from: www.micromedexsolutions.com
  • 18. Ellis K, Grindrod K, Tully S, McFarlane T, Chan KKW, Wong WWL. Understanding the feasibility of implementing CAR T-cell therapies from a Canadian perspective. Healthc Policy. 2021;16(3):89–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Caro J, Möller J, Karnon J, Stahl J, Ishak J. Discrete Event Simulation for Health Technology Assessment. New York: CRC Press; 2016. [Google Scholar]
  • 20. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Howlader N, Mariotto AB, Besson C, et al. Cancer-specific mortality, cure fraction, and noncancer causes of death among diffuse large B-cell lymphoma patients in the immunochemotherapy era. Cancer. 2017;123(17):3326–34. [DOI] [PubMed] [Google Scholar]
  • 22. Corbett MS, Duarte AI, Walker SM, Wright K, Simmonds MC, Palmer SJ. Tisagenlecleucel for Treating Relapsed or Refractory Diffuse Large B-cell Lymphoma: Single Technology Appraisal Evidence Assessment Group Report. London: National Institute for Health and Care Excellence; 2018. [Google Scholar]
  • 23. Canadian Agency for Drugs and Technologies in Health. Guidelines for the Economic Evaluation of Health Technologies: Canada. Ottawa (Canada): Canadian Agency for Drugs and Technologies in Health; 2017. [Google Scholar]
  • 24. Ellis K. Cost-effectiveness of Chimeric Antigen Receptor T-cell Therapy for Treating Large B-cell Lymphoma Patients in Canada. Waterloo (Canada): University of Waterloo, 2020. [Google Scholar]
  • 25. Institute for Clinical and Economic Review. Chimeric Antigen Receptor T-cell Therapy for B-cell Cancers: Effectiveness and Value Final Evidence Report. Boston: Institute for Clinical and Economic Review; 2018. [Google Scholar]
  • 26. Feugier P, Van Hoof A, Sebban C, et al. Long-term results of the R-CHOP study in the treatment of elderly patients with diffuse large B-cell lymphoma: a study by the Groupe d’Etude des Lymphomes de l’Adulte. J Clin Oncol. 2005;23(18):4117–26. [DOI] [PubMed] [Google Scholar]
  • 27. Friedberg JW. Relapsed/refractory diffuse large B-cell lymphoma. Hematology Am Soc Hematol Educ Program. 2011;2011:498–505. [DOI] [PubMed] [Google Scholar]
  • 28. Guyot P, Ades AE, Ouwens MJ, Welton NJ. Enhanced secondary analysis of survival data: reconstructing the data from published Kaplan-Meier survival curves. BMC Med Res Methodol. 2012;12:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Gibson E, Koblbauer I, Begum N, et al. Modelling the survival outcomes of immuno-oncology drugs in economic evaluations: a systematic approach to data analysis and extrapolation. Pharmacoeconomics. 2017;35(12):1257–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Statistique Canada. Life tables, Canada, provinces and territories. 2021. Available from: https://www150.statcan.gc.ca/n1/en/catalogue/84-537-X. [Accessed 2022].
  • 31. 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):3490–6. [DOI] [PubMed] [Google Scholar]
  • 32. Ontario Health. Ontario Case Costing Initiative: Cost Analysis Tool. Toronto (Canada): Ontario Health; 2018. [Google Scholar]
  • 33. Ontario. Schedule of Benefits: Physician Services under the Health Insurance Act. Toronto (Canada): Ministry of Health; 2022. [Google Scholar]
  • 34. Ontario. Ontario Drug Benefit: Formulary Search. Toronto (Canada): Ontario Health; 2022. [Google Scholar]
  • 35. Canadian Institute for Health Information. 2017 Hospital per Diem Rates Using 2015-16 Canadian MIS Database (CMDB) Data. Ottawa (Canada): Canadian Institute for Health Information; 2017. [Google Scholar]
  • 36. Ontario. Interprovincial Billing Rates for Designated High Cost Transplants Effective for Discharges on or after April 1, 2020. Toronto (Canada): Ontario Health; 2020. [Google Scholar]
  • 37. Post-treatment monitoring frequency discussion with clinician. Toronto (Canada); 2021. [Google Scholar]
  • 38. Hayden PJ, Sirait T, Koster L, Snowden JA, Yakoub-Agha I. An international survey on the management of patients receiving CAR T-cell therapy for haematological malignancies on behalf of the Chronic Malignancies Working Party of EBMT. Curr Res Transl Med. 2019;67(3):79–88. [DOI] [PubMed] [Google Scholar]
  • 39. de Oliveira C, Pataky R, Bremner KE, et al. Phase-specific and lifetime costs of cancer care in Ontario, Canada. BMC Cancer. 2016;16(1):809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Tanuseputro P, Wodchis WP, Fowler R, et al. The health care cost of dying: a population-based retrospective cohort study of the last year of life in Ontario, Canada. PLoS One. 2015;10(3):e0121759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Statistics Canada. Consumer Price Index, by province. 2022. Available from: https://www.statcan.gc.ca/en/subjects-start/prices_and_price_indexes/consumer_price_indexes [Google Scholar]
  • 42. Organisation for Economic Co-operation and Development. Purchasing power parities (PPP). 2022. Available from: https://data.oecd.org/conversion/purchasing-power-parities-ppp.htm [Accessed August, 2022]. [Google Scholar]
  • 43. Lin VW, Jiang Y, Chuang LH, Navale L, Cheng P, Purdum A. Health utilities for patients with relapsed or refractory large B-cell lymphoma (R/R-LBCL): ad hoc analysis from an axicabtagene ciloleucel (Axi-cel) safety management study. Bone Marrow Transplant. 2019;53:878–87. [Google Scholar]
  • 44. Doorduijn JK, van der Holt B, van Imhoff GW, et al. CHOP compared with CHOP plus granulocyte colony-stimulating factor in elderly patients with aggressive non-Hodgkin’s lymphoma. J Clin Oncol. 2003;21(16):3041–50. [DOI] [PubMed] [Google Scholar]
  • 45. Sung L, Buckstein R, Doyle JJ, Crump M, Detsky AS. Treatment options for patients with acute myeloid leukemia with a matched sibling donor: a decision analysis. Cancer. 2003;97(3):592–600. [DOI] [PubMed] [Google Scholar]
  • 46. Patrick DL, Powers A, Jun MP, et al. Effect of lisocabtagene maraleucel on HRQoL and symptom severity in relapsed/refractory large B-cell lymphoma. Blood Adv. 2021;5(8):2245–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Maurer MJ, Ghesquières H, Jais JP, et al. Event-free survival at 24 months is a robust end point for disease-related outcome in diffuse large B-cell lymphoma treated with immunochemotherapy. J Clin Oncol. 2014;32(10):1066–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. 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):2531–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Canadian Agency for Drugs and Technologies in Health. Axicabtagene Ciloleucel for Diffuse Large B-cell Lymphoma: Economic Review Report. Ottawa (Canada): Canadian Agency for Drugs and Technologies in Health; 2019. [PubMed] [Google Scholar]
  • 50. Cher BP, Gan KY, Aziz MIA, et al. Cost utility analysis of tisagenlecleucel vs salvage chemotherapy in the treatment of relapsed/refractory diffuse large B-cell lymphoma from Singapore’s healthcare system perspective. J Med Econ. 2020;23(11):1321–9. [DOI] [PubMed] [Google Scholar]
  • 51. Lin JK, Muffly LS, Spinner MA, Barnes JI, Owens DK, Goldhaber-Fiebert JD. Cost effectiveness of chimeric antigen receptor T-cell therapy in multiply relapsed or refractory adult large B-cell lymphoma. J Clin Oncol. 2019;37(24):2105–19. [DOI] [PubMed] [Google Scholar]
  • 52. Qi CZ, Bollu V, Yang H, Dalal A, Zhang S, Zhang J. Cost-effectiveness analysis of tisagenlecleucel for the treatment of patients with relapsed or refractory diffuse large B-cell lymphoma in the United States. Clin Ther. 2021;43(8):1300–1319.e8. [DOI] [PubMed] [Google Scholar]
  • 53. Wakase S, Teshima T, Zhang J, et al. Cost effectiveness analysis of tisagenlecleucel for the treatment of adult patients with relapsed or refractory diffuse large B cell lymphoma in Japan. Transplant Cell Ther. 2021;27(6):506.e1–506.e10. [DOI] [PubMed] [Google Scholar]
  • 54. Wang XJ, Wang YH, Li SCT, et al. Cost-effectiveness and budget impact analyses of tisagenlecleucel in adult patients with relapsed or refractory diffuse large B-cell lymphoma from Singapore’s private insurance payer’s perspective. J Med Econ. 2021;24(1):637–53. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

sj-docx-1-mdm-10.1177_0272989X241234070 – Supplemental material for CAR T-cell Therapy for Diffuse Large B-cell Lymphoma in Canada: A Cost-Utility Analysis

Supplemental material, sj-docx-1-mdm-10.1177_0272989X241234070 for CAR T-cell Therapy for Diffuse Large B-cell Lymphoma in Canada: A Cost-Utility Analysis by Lisa Masucci, Feng Tian, Stephen Tully, Zeny Feng, Tom McFarlane, Kelvin K. W. Chan and William W. L. Wong in Medical Decision Making


Articles from Medical Decision Making are provided here courtesy of SAGE Publications

RESOURCES