CADTH Reimbursement Review

Axicabtagene Ciloleucel (Yescarta)

Sponsor: Gilead Sciences Canada Inc.

Therapeutic area: Relapsed or refractory follicular lymphoma

This multi-part report includes:

Clinical Review

Pharmacoeconomic Review

Ethics Review

Stakeholder Input

Clinical Review

Abbreviations

AE

adverse event

allo-SCT

allogeneic stem cell transplant

auto-SCT

autologous stem cell transplant

CAR

chimeric antigen receptor

CI

confidence interval

CR

complete response

CRR

complete response rate

CRS

cytokine release syndrome

DLBCL

diffuse large B-cell lymphoma

DOR

duration of response

ECOG PS

Eastern Cooperative Oncology Group Performance Status

FAS

full analysis set

FL

follicular lymphoma

FLIPI

Follicular Lymphoma Internal Prognostic Index

GELF

Groupe d’Étude des Lymphomes Folliculaires

HR

hazard ratio

HRQoL

health-related quality of life

KM

Kaplan-Meier

LC

Lymphoma Canada

MZL

marginal zone lymphoma

NE

not evaluable

NHL

non-Hodgkin lymphoma

OH-CCO

Ontario Health-Cancer Care Ontario

ORR

objective response rate

OS

overall survival

OR

odds ratio

PFS

progression-free survival

PI3K

phosphoinositide 3-kinase

POD24

progression of disease within 24 months

r/r

relapsed or refractory

SAE

serious adverse event

SCT

stem cell transplant

SD

standard deviation

SMR

standardized mortality ratio

TEAE

treatment-emergent adverse event

TTNT

time to next treatment

Executive Summary

An overview of the submission details for the drug under review is provided in Table 1.

Table 1: Background Information of Application Submitted for Review

Item

Description

Drug product

Axicabtagene ciloleucel (Yescarta); cell suspension in patient-specific single-infusion bag, for IV infusion

Sponsor

Gilead Sciences Canada Inc.

Indication

For the treatment of adult patients with relapsed or refractory grade 1, 2, or 3a follicular lymphoma (FL) after 2 or more lines of systemic therapy.

Reimbursement request

As per indication

Health Canada approval status

NOC/c

Health Canada review pathway

Standard review

NOC date

September 28, 2022

Recommended dose

A single-dose, 1-time treatment; target dose of 2 × 106 CAR-positive T cells/kg body weight (range, 1 × 106 cells/kg to 2.4 × 106 cells/kg) to a maximum of 2 × 108 CAR-positive viable T cells for patients weighing ≥ 100 kg

CAR = chimeric antigen receptor; NOC/c = Notice of Compliance with conditions.

Introduction

Non-Hodgkin lymphoma (NHL) encompasses a heterogenous group of more than 80 closely related cancers.1 It is characterized by the abnormal and uncontrolled proliferation of cells (i.e., T cells, B cells, and natural killer cells) of the lymphatic system.2,3 Follicular lymphoma (FL), a subtype of NHL, is an indolent B-cell lymphoma originating from the germinal centre of lymphoid tissues2-6 and characterized by slow growth and spread.7 It makes up 20% to 30% of all NHL cases.1 The sponsor-calculated overall incidence rate of FL in Canada (based on the NHL age-standardized incidence rates [25.7 per 100,000] and the proportion of FL among NHL cases [25%])4,6 reported was 7.21 per 100,000.4,8,9 Although responsive to initial first- or second-line therapies, FL is characterized by a relapsing and remitting disease course, especially in advanced disease stages. Patients will eventually require multiple treatments to manage or slow disease progression throughout their lifetime as response to treatments decline upon repeated therapy.3,10-12 The clinical experts consulted by the sponsor reported that approximately 30.95% of incident FL patients would progress to third-line treatment, of whom 60% would proceed to receive active therapy.13-15 FL can be further classified into 3 grades (1, 2, and 3 [a and b]) based on cell structures under the microscope, specifically, the number of large FL cells (centroblasts) observed.16 Grades 1, 2, and 3a diseases are generally considered low grade or slow growing compared to grade 3b, which grows fast and is considered high-grade lymphoma. According to the Canadian Cancer Society, 91% of patients considered “low risk” at diagnosis as per the Follicular Lymphoma Internal Prognostic Index (FLIPI) score have a 5-year survival rate and 71% have a 10-year survival rate; 78% of patients considered “intermediate risk” have a 5-year survival rate and 51% have a 10-year survival rate; and 53% of patients considered “high risk” have a 5-year survival rate and 36% have a 10-year survival rate.17

The objective of this report is to review and critically appraise the evidence submitted by the sponsor on the beneficial and harmful effects of axicabtagene ciloleucel, at a target dose of 2 × 106 chimeric antigen receptor (CAR)-positive T cells/kg body weight, to a maximum of 2 × 108 CAR-positive viable T cells, by IV infusion, in the treatment of adult patients with relapsed or refractory (r/r) FL after 2 or more lines of systemic therapy.

Stakeholder Perspectives

The information in this section is a summary of input provided by the patient and clinician groups who responded to CADTH’s call for input and from clinical expert(s) consulted by CADTH for the purpose of this review.

Patient Input

One patient advocacy group, Lymphoma Canada (LC), provided input for this review. LC is a national Canadian registered charity whose mission is to empower patients and the lymphoma community through education, support, advocacy, and research. The LC patient group expressed the need for accessible treatment options for patients, emphasizing that local access to treatments would significantly improve patients' quality of life and experience by reducing fear and the risk of getting sick while travelling.

LC gathered information for this input via online surveys completed anonymously by patients between April 21, 2022, and April 3, 2023. Of the 143 responses submitted, 3 respondents reported prior experience with axicabtagene ciloleucel. Respondents indicated that fatigue (50%), body aches and pain (33%), enlarged lymph nodes (33%), indigestion (32%), and bodily swelling (21%) were the most challenging symptoms that impacted their quality of life at the time of diagnosis. Respondents described FL symptoms as challenges in their daily lives that impacted their ability to travel (46%), spend time with family or friends (41%), exercise (37%), concentrate (36%), and work or complete school or volunteer activities (35%) . About half (49%) of the respondents reported that they went through a period of “watchful waiting” before commencing treatment. Most respondents (43%) had received 1 line of treatment. The most common treatments reported by respondents who had received 1 or 2 lines of systemic therapy included chemotherapy, chemoimmunotherapy, rituximab with or without bendamustine, or radiation. The posttreatment symptoms that most significantly negatively impacted respondents included treatment-related fatigue (28%), immediate side effects of treatment (26%), and low activity level (23%). Fatigue (69%), hair loss (41%), and constipation (38%) were the most common side effects reported by respondents. The most important outcomes highlighted by respondents included long life (84%), longer disease remission (82%), improved quality of life and ability to perform daily activities (69%), ability to control disease symptoms (63%), and ability to normalize blood counts (58%). Two-thirds of the respondents indicated that they were willing to tolerate nonsevere side effects for a short-term as a trade-off for a novel treatment. Two respondents who had previously received axicabtagene ciloleucel reported having access to the drug via a clinical trial. Reported side effects were cytokine release syndrome (CRS), neutropenia, febrile neutropenia, thrombocytopenia, constipation, and swelling. Some of the challenges the respondents associated with receiving axicabtagene ciloleucel included the frequent monitoring of side effects postinfusion, the inability to perform daily activities, and being away from family and friends. Both respondents said that they had a good or very good experience with axicabtagene ciloleucel and would recommend this treatment to other patients with r/r FL.

Clinician Input

Input From Clinical Experts Consulted by CADTH

A panel of 4 experts with experience treating r/r FL were consulted to determine the unmet needs, place in therapy, the patient population identified as most and least likely to benefit from treatment, when to start treatment, how best to assess response to treatment, and guidance for discontinuing treatment. The clinical experts indicated that the most important goals for treatment are to prolong life, and that the greatest unmet needs exist in patients with cancer that progresses within 2 years after their initial therapy, the patients who have already received autologous stem cell transplant (auto-SCT) or are ineligible for auto-SCT, or those who have been double refractory to earlier line treatments (implying limited treatment options available to them). The clinical panel suggested that axicabtagene ciloleucel be used as third- or later-line treatments for patients with r/r FL. These patients usually have a treatment response that lasts less than 6 months after their last treatment (medication or SCT).

The clinical panel indicated that, in practice, CAR T-cell therapy is used in a patient population that is broader than the population selected and recruited for clinical trials. The panel indicated that in clinical practice, patients are evaluated and followed in a manner similar to that described in the clinical trials of FL treatments. Remission and survival are measured. Physical exams and imaging exams are routinely conducted to assess the patient’s response to CAR T-cell therapy. The panel suggested that meaningful responses to treatment with axicabtagene ciloleucel would include a high complete remission rate, durability of treatment response, and long-term progression-free survival (PFS) and overall survival (OS). The panel indicated that in the event of treatment failure after infusion with axicabtagene ciloleucel, patients may participate in a clinical trial. In the absence of clinical trial, they may try a different chemoimmunotherapy that they have not been exposed to or undergo auto-SCT if they have not already received this therapy. The panel emphasized the importance of an accredited multidisciplinary team involving hematologists, infectious disease specialists, neurologists, an intensive care unit team, and other specialists to diagnose, treat, and monitor the patients receiving axicabtagene ciloleucel and to ensure the safe and effective delivery of this treatment.

Clinician Group Input

Input from 1 clinician group, the Ontario Health — Cancer Care Ontario (OH-CCO) Hematology Cancer Drug Advisory Committee, was summarized for this review. The disease course of FL varies for every patient. Some patients may present with long remissions between therapies while others would have refractory disease. Current treatment goals for patients with FL, according to the clinician group include palliative care and, in some scenarios, treatment with curative intent using allogeneic stem cell transplant (allo-SCT). The most important goals outlined were to delay disease progression, improve patient health-related quality of life, and alleviate symptoms. The OH-CCO Hematology Cancer Drug Advisory Committee acknowledged that current treatment options do not meet the needs of patients with r/r FL. The clinicians in the committee mentioned that patients who become refractory to chemotherapy have no other treatment options to delay the disease. In addition, the clinicians highlighted that repeated administration of cytotoxic therapy may be associated with marrow damage (myelodysplastic syndrome), which further limits the ability to treat patients, and adversely affects quality of life. Hence, there is a need for treatment options that patients can tolerate. Treatment with CAR T-cell therapy, according to the committee members, is not anticipated to cause long-term marrow damage issues. The clinicians noted that a third-line therapy with a CAR T-cell therapy would be appropriate, given that current therapy provides less benefit to patients with r/r FL disease. Patients eligible to receive axicabtagene ciloleucel in clinical practice would be similar to patients included in the clinical trial, according to the experts. However, patients with severe organ dysfunction, poor performance status, and uncontrolled infections would be ineligible. The clinicians pointed out that patients who had received prior CD19-directed therapy should be considered for treatment with CAR T-cell therapy and highlighted the need for flexibility around patients’ Eastern Cooperative Oncology Performance Status (ECOG PS) or Karnofsky Performance Status (KPS) scores. The committee members noted that some patients might become ineligible to receive CAR T-cell therapy during manufacturing, which may lead to discontinuation.

Drug Program Input

The drug programs provide input on each drug being reviewed through CADTH’s Reimbursement Review processes by identifying issues that may impact their ability to implement a recommendation. The drug plans identified implementation issues related to initiation, prescribing, generalizability, funding algorithm, care provision, system issues, and economic considerations. The clinical experts consulted by CADTH for this review weighed evidence from the included study and other clinical considerations to provide responses to the drug plan’s implementation questions.

Clinical Evidence

Systematic Review

Description of Studies

ZUMA-5 is a multicentre, international, open-label, single-arm phase II trial.18 The study objective was to determine the efficacy and safety of axicabtagene ciloleucel in patients with r/r FL or marginal zone lymphoma (MZL) after 2 or more lines of systemic therapy. Between |||| |||| ||| |||| ||||, 127 FL patients were enrolled at 15 sites in the US and 2 in France. There were no study sites in Canada. Prior to receiving any treatments, patients underwent leukapheresis to obtain T cells as part of the manufacturing process for axicabtagene ciloleucel. Patients were then treated with cyclophosphamide and fludarabine lymphodepleting chemotherapy between 5 and 3 days before axicabtagene ciloleucel infusion. After 2 days of rest, patients received axicabtagene ciloleucel through IV infusion with a target dose of 2 × 106 anti-CD19 CAR T cells/kg body weight. Analyses were conducted at 18 months, 24 months, and 36 months. The statistical analysis plan prespecified that tests on the inferential analysis set be conducted at 18 months, that is, on the date when 80 patients had been followed for at least 18 months. Using all enrolled patients, analyses were conducted at 18 months, 24 months (not presented), and 36 months. The data cut-off for the 18-month analysis was September 14, 2020; the data cut-off for the 36-month analysis was March 31, 2022.

At the 36-month time point for analysis, the median age was 60 years (range, 34 years to 79 years) and 62% of patients had an ECOG PS score of 0. Of the enrolled patients, 69% were refractory, defined as progressing within 6 months of their most recent treatment. Most patients enrolled in ZUMA-5 had received 2 prior therapies |||||, 26% had received 3 prior therapies, 20% had received 4 prior therapies, and 17% had received 5 or more prior therapies. The proportion of patients who had received a prior auto-SCT was 24%, while the proportion of patients with high bulk tumour was 51%. The proportion of patients who had progressed within 24 months of anti-CD20 chemotherapy combination therapy (i.e., progression of disease within 24 months [POD24]) was 55%.

Efficacy Results

A summary of the efficacy results in the ZUMA-5 trial is shown in Table 2.

Overall Survival

The proportion of patients who had died due to any cause was ||| after 36 months of follow-up. The median OS had not been reached. Clinical experts considered OS to be the ideal survival end point for decision-making, but acknowledged that due to the extended survival periods seen in r/r FL, immature OS results are common. The Kaplan-Meier (KM) survival probability at 18 months was ||||| ||||| || |||||, at 24 months was ||||| ||||| || |||||, and at 36 months was 75.5% (95% confidence interval [CI], 66.9% to 82.2%).

Progression-Free Survival

The proportion of patients who experienced a progression event was ||| after 36 months of follow-up. The median PFS was 40.2 months (95% CI, 28.9 months to not evaluable [NE]). The KM PFS probability at 18 months was ||||| ||||| || |||||, at 24 months was ||||||||| ||| ||||| ||||| || |||||, and at 36 months was 54.4% (95% CI, 44.2% to 63.5%).

Objective Response Rate

At the 36-month time point, the estimated objective response rate (ORR) as per investigator assessment was a clinically meaningful 94% (95% CI, 88% to 97%) in the full analysis set (FAS), while the complete response rate (CRR) was 79% ||| || |||. According to clinical experts, and within the context of the extended survival periods in r/r FL, ORR and CRR are considered acceptable surrogate end points for more important survival end points.

The primary end point in the ZUMA-5 trial was ORR at the 18-month analysis in the inferential analysis set, with a prespecified threshold of 40% for ORR and 15% for CRR. The estimated ORR as per central assessment in the 18-month inferential analysis set was 94% ||| || |||| | ||||| ||||||| and the CRR was 79% ||| || |||| | ||||| |||||||. Subgroup analyses conducted on prespecified baseline characteristics were consistent with the overall results.

Duration of Response

At the 36-month time point for analysis, ||| of patients with a response had experienced a loss-of-response event. The estimated median duration of response (DOR) was 38.6 months (95% CI, 29.0 months to NE), which was considered clinically meaningful by the clinical experts consulted by CADTH. The KM-estimated event-free probability at 18 months was ||||| ||||| || |||||, at 24 months was ||||| ||||| || |||||, and at 36 months was ||||| ||||| || |||||.

Time to Next Treatment

At the 36-month time point for analysis, ||| of patients had experienced a time-to-next-treatment (TTNT) event. The median TTNT was NE months (95% CI, 37.8 months to NE). The KM-estimated event-free probability at 18 months was ||||| ||||| || |||||, at 24 months was ||||| ||||| || |||||, and at 36 months was 59.5% (95% CI, 50.2% to 67.6%).

Harms Results

At the 36-month time point for analysis, a total of ||| of patients in the safety analysis set experienced a treatment-emergent adverse event (TEAE) with pyrexia |||||, hypotension |||||, headache |||||, and fatigue ||||| the most frequently reported TEAEs. A total of ||| of patients in the safety analysis experienced a serious adverse event (SAE), with pyrexia ||||| and pneumonia |||| the most frequently reported SAEs. At the 36-month time point, ||| of patients in the safety analysis set had died. The most common reason for death was progressive disease ||||, following by an adverse event (AE) due to reasons other than progressive disease or subsequent therapy |||| and secondary malignancy ||||.

Notable harms identified included CRS, neurologic events, cytopenias, infection, and hypogammaglobulinemia. At the 36-month analysis ||| of patients in the safety analysis set had experienced CRS, with || experiencing a grade 3 or higher CRS. Neurologic events were reported in ||| of patients, with ||| reporting a grade 3 or higher neurologic event. Cytopenias were reported in ||| of patients, with ||| of patients reporting a grade 3 or higher cytopenia. Infections were reported in ||| of patients, with ||| reporting a grade 3 or higher infection. Hypogammaglobulinemia was reported in ||| of patients, with || of patients reporting a grade 3 or higher hypogammaglobulinemia.

Table 2: Summary of Key Results From Pivotal Studies and RCT Evidence

Measure

ZUMA-5

Inferential analysis set

18 months

(N = 86)

FAS

18 months

(N = 127)

FAS

36 months

(N = 127)

OS

Number of patients with event, n (%)

|| ||||

|| ||||

|| ||||

OS time (months), median (95% CI)

NE (31.6 to NE)

|| |||| |||

NE (NE to NE)

PFS

Number of patients with event, n (%)

|| ||||

|| ||||

|| ||||

PFS time (months), median (95% CI)

NE (23.5 to NE)

|| |||| |||

40.2 (28.9 to NE)

Response

ORR, n (% [95% CI])

81 (94 [||| || ||||])

||| ||| ||| || ||||

119 (94 [88 to 97])

CRR, n (% [95% CI])

68 (79 [||| || ||||])

|| ||| ||| || |||||

100 (79 [||| || |||])

DOR

| | ||

| | |||

N = 119

Number of patients with events, n (%)

|| ||||

|| ||||

|| ||||

DOR time (months), median (95% CI)a

NE (NE to NE)

|| |||| |||

38.6 (29.0 to NE)

TTNT

Number of patients with events, n (%)

|| ||||

|| ||||

|| ||||

TTNT time (months), median (95% CI)a

NE (NE to NE)

|| |||| |||

NE (37.8 to NE)

Harms, n (%)

N

NA

| | |||

| | |||

Patients with any TEAE

NA

||| ||||

||| ||||

Patients with any SAE

NA

|| ||||

|| ||||

Deaths

NA

|| ||||

|| ||||

AESI: CRS

NA

|| ||||

|| ||||

AESI: CRS grade ≥ 3

NA

| |||

| |||

AESI: Neurologic event

NA

|| ||||

|| ||||

AESI: Neurologic event grade ≥ 3

NA

|| ||||

|| ||||

AESI: Cytopenias

NA

|| ||||

|| ||||

AESI: Cytopenias grade ≥ 3

NA

|| ||||

|| ||||

AESI: Infection

NA

|| ||||

|| ||||

AESI: Infection grade ≥ 3

NA

|| ||||

|| ||||

AESI: Hypogammaglobulinemia

NA

|| ||||

|| ||||

AESI: Hypogammaglobulinemia grade ≥ 3

NA

| |||

| |||

AESI = adverse event of special interest; CI = confidence interval; CRR = complete response rate; CRS = cytokine release syndrome; DOR = duration of response; FAS = full analysis set; NA = not applicable; NE = not evaluable; ORR = objective response rate; OS = overall survival; PFS = progression-free survival; RCT = randomized controlled trial; SAE = serious adverse event; TEAE = treatment-emergent adverse event; TTNT = time to next treatment.

Note: ORR, CRR, PFS, and DOR are reported according to central assessment at the 18-month analysis and as per investigator assessment for the 36-month analysis.

Source: ZUMA-5 Clinical Study Report.18

Critical Appraisal

The ZUMA-5 trial, the only eligible study identified by the sponsor, was a phase II, single-arm, open-label clinical trial. The lack of comparative data is a key limitation to the interpretation of the results from a single-arm trial, as it is difficult to distinguish between the effect of the intervention, a placebo effect, or the effect of natural history. Due to the open-label design of the trial, the response outcomes measures (i.e., ORR, CRR, DOR, PFS) and subjective harms are at risk of measurement or reporting bias, though the direction of this bias is unclear. It is noted that these limitations are partly addressed through the use of a prespecified threshold for ORR and CRR end points and the use of central assessment.

Another important limitation of the ZUMA-5 trial is related to the insufficient follow-up time to draw strong conclusions on the long-term survival impacts of axicabtagene ciloleucel for patients with r/r FL. The clinical experts consulted by CADTH noted that r/r FL is a disease that can have very long periods of PFS and survival, suggesting that the follow-up duration was not long enough to fully capture the effects on OS and PFS. In addition, subsequent treatments could confound the long-term survival results of the ZUMA-5 trial.

According to the clinical experts consulted by CADTH, the ZUMA-5 trial patient population is overall representative of the patients in the population with r/r FL in Canada who would be receiving axicabtagene ciloleucel. However, the clinical experts noted that patients seen in clinical practice would include those with poorer performance status (the ZUMA-5 trial only included patients with an ECOG PS score of 0 or 1, whereas clinical experts suggest that an ECOG PS score of 2 may be treated in the clinical setting), and patients with more comorbidities. The clinical experts had different opinions regarding patients who received prior CD19-targeted therapy; some suggested that any prior CD19-targeted therapy would preclude the use of axicabtagene ciloleucel, whereas others suggested that only patients who are refractory to CD19-targeted therapy (did not respond or relapsed within 6 months) would not be suitable candidates for treatment with axicabtagene ciloleucel. According to the clinical experts consulted by CADTH, the efficacy outcomes used in this study are clinically relevant and important for the clinical trials in r/r FL, with the notable exception of health-related quality of life (HRQoL) outcomes, which are important to patients but were excluded from the ZUMA-5 trial. As such, it is not possible to determine how the introduction of axicabtagene ciloleucel will impact the HRQoL of patients in Canada.

Long-Term Extension Studies

No long-term extension studies were submitted as part of this review.

Indirect Comparisons

No indirect treatment comparisons were submitted as part of this review.

Studies Addressing Gaps in the Evidence From the Systematic Review

The aim of the sponsor was to provide an estimate of relative efficacy against standard of care therapies in patients with r/r FL who have received 2 or more lines of therapy.19

Description of Studies

The relative efficacy of axicabtagene ciloleucel versus standard of care estimated among the ZUMA-5 treated population using propensity scores with standardized mortality ratio (SMR) weights. The SCHOLAR-5 trial, the standard of care cohort, is a retrospective, observational, multicentre, database study of patients with r/r FL (grades 1 to 3a) who have received 2 or more systemic therapies. Patient-level data for the ZUMA-5 and SCHOLAR-5 trials were used to inform the comparative analysis. Propensity scores were calculated for each patient in the pooled analysis set to account for differences in baseline characteristics across populations. Selection of variables for the propensity score model was determined in a hierarchal manner and based on the advice of investigators/clinical experts with the goal of minimizing the imbalance in prognostically important covariates.

Efficacy Results

The ORR in the ZUMA-5 population was 93.7% compared to 54.0% in the propensity score–weighted SCHOLAR-5 population (odds ratio [OR] = 12.66; 95% CI, 5.24 to 30.57). The CRR in the ZUMA-5 population was 78.7% compared to 34.9% in the propensity score–weighted SCHOLAR-5 population (OR = 6.90; 95% CI, 3.62 to 13.18). The median DOR in the ZUMA-5 population was 38.64 months (95% CI, 29.04 months to NE) compared to ||||| |||||| ||||| || |||||| in the propensity score–weighted SCHOLAR-5 population (hazard ratio [HR] = |||| ||||| || |||||).

The median PFS in the ZUMA-5 population was 40.21 months (95% CI, 28.94 months to NE) compared to 12.97 months (95% CI, 7.75 months to 15.47 months) in the propensity score–weighted SCHOLAR-5 population (HR = 0.27; 95% CI, 0.18 to 0.41). The median OS in the ZUMA-5 population was NE (95% CI, NE to NE) compared to NE (38.40 months to NE) in the propensity score–weighted SCHOLAR-5 population (HR = 0.56; 95% CI, 0.33 to 0.95). The median TTNT in the ZUMA-5 population was NE (95% CI, 37.85 months to NE) compared to 26.61 months (95% CI, 12.65 months to NE) in the propensity score–weighted SCHOLAR-5 population, with HR of 0.60 (95% CI, 0.39 to 0.93).

Harms Results

Safety end points were not included in the analysis.

Critical Appraisal

Due to differences between the ZUMA-5 and SCHOLAR-5 cohorts in treatment allocation, it is possible that the treatment effect estimate is confounded by imbalances in prognostic covariates across populations. The sponsor identified and adjusted for several important variables, resulting in a suitable balance of these characteristics across both populations; however, important characteristics such as FLIPI score could not be adjusted for due to missing data. Characteristics such as ECOG PS, FL grade, and whether patients were double refractory differed significantly between populations after propensity score weighting. The clinical experts consulted by CADTH suggested that differences in ECOG PS score and the proportion of patients who are double refractory could impact how patients respond to treatment. The direction of this impact is uncertain, with some differences (e.g., double refractory status and FL grade) potentially favouring the SCHOLAR-5 comparator over axicabtagene ciloleucel, and some differences (e.g., ECOG PS score) potentially favouring axicabtagene ciloleucel over the SCHOLAR-5 comparator.

There is additional uncertainty in the results due to the low effective sample sizes in both the ZUMA-5 trial and the SCHOLAR-5 cohort. The removal of the DELTA patients from the SCHOLAR-5 cohort resulted in a statistically significant change in the mean number (standard deviation [SD]) of lines of prior therapy: |||| |||||| in the SCHOLAR-5 trial compared to |||| |||||| in the ZUMA-5 trial. Differences between populations in the number of lines of prior therapy in particular affect determining how patients would be expected to respond to treatment. The proportion of patients with POD24 and the proportion of patients who were refractory to their most recent treatment was also reduced with the exclusion of the DELTA cohort, indicating that their removal from the SCHOLAR-5 cohort resulted in a population with a lower risk prognosis.

Conclusions

Evidence from a single-arm study (the ZUMA-5 trial) suggests that treatment with axicabtagene ciloleucel affects clinically important tumour responses, including complete remission, in adult patients with r/r FL after 2 or more lines of systemic therapies. Due to the single-arm design of the trial and limited duration of follow-up, there is insufficient evidence to determine the magnitude of the effect of axicabtagene ciloleucel on OS and PFS. HRQoL outcomes were not included in the ZUMA-5 trial and therefore the impact of axicabtagene ciloleucel on patients HRQoL is unknown. The harms associated with the axicabtagene infusion are as expected given the mechanism of action and prior experience in other indications. The comparison of the ZUMA-5 trial to the retrospective SCHOLAR-5 external control was limited by heterogeneity across study designs and populations. Specifically, the inability to adjust for ECOG PS and double refractory status can introduce bias to the estimation procedure within the comparative populations. Generalizability to individuals that do not meet the ZUMA-5 trial criteria is also in question. Therefore, the magnitude of the comparative efficacy estimates for axicabtagene ciloleucel against standard of care in the Canadian setting is uncertain.

Introduction

The objective of this report is to review and critically appraise the evidence submitted by the sponsor on the beneficial and harmful effects of axicabtagene ciloleucel (target of 2 × 106 CAR-positive viable T cells/kg body weight, for IV use; range, 1 × 106 cells/kg to 2.4 × 106 cells/kg) in the treatment of adult patients with r/r grade 1, 2, or 3a FL after 2 or more lines of systemic therapy.

Disease Background

Contents within this section have been informed by materials submitted by the sponsor and clinical expert input. The following have been summarized and validated by the CADTH review team.

NHL encompasses a heterogenous group of more than 80 closely related cancers.1 It is characterized by the abnormal and uncontrolled proliferation of cells (i.e., T cells, B cells, and natural killer cells) of the lymphatic system.2,3 About 85% to 90% of NHLs develop from B lymphocytes, and the rest from T lymphocytes or natural killer cells.2,20 NHL is the fifth most common cancer diagnosed in Canada, and is commonly diagnosed in adults aged 50 years or older.1 In 2022, an estimated 11,400 new diagnoses of NHL and 3,000 deaths from NHL were projected for Canada.21 The estimates were higher in males (6,600 new cases and 1,700 deaths) than in females (4,800 new cases and 1,250 deaths).21 Risk factors for all NHL-related cancers include immune disorders (e.g., rheumatoid arthritis, Sjögren syndrome, systemic lupus erythematosus), the use of immunosuppressive therapies, bacterial and viral infections (e.g., Helicobacter pylori, Epstein-Barr virus, and hepatitis C virus), family history and genetics, and occupational and lifestyle risk factors.1-3,20 NHL can affect any organ in the body and has a wide range of clinical presentations.3,20

FL, a subtype of NHL, is an indolent B-cell lymphoma originating from the germinal centre of lymphoid tissues,2-6 and characterized by slow growth and spread.7 FL makes up 20% to 30% of all NHL cases1 and up to 70% of all indolent NHL cases. There are limited epidemiological data on FL in Canada compared with other NHL-associated cancers. One study that looked at trends (incidence and mortality) of FL across Canada using data from 3 registries (the Canadian Cancer Registry, the Registre québécois du cancer, and the Canadian Vital Statistics database) reported that there were about 22,625 new cases of FL between 1992 and 2010. The mean age of patients was 60.8 years at diagnosis, with equal incidence rates observed in males and females (50% in each sex) in the study population.6 The authors also reported a variability in incidence rates across provinces, with rates in Manitoba, New Brunswick, Nova Scotia, and Prince Edward Island notably higher than the national average.6,22 The sponsor-calculated overall incidence rate of FL in Canada (based on the age-standardized incidence rates of NHL [25.7 per 100,000] and the reported proportion of FL among NHL cases [25%])4,6 was 7.21 per 100,000.4,8,9

Although responsive to initial first- or second-line therapies, FL is characterized by a relapsing and remitting disease course, especially in advanced disease stages. Patients will eventually require multiple treatments to manage or slow disease progression throughout their lifetime as response to treatments declines with repeated therapy.3,10-12 The clinical experts consulted by the sponsor reported that approximately |||||| of incident FL patients would progress to third-line treatment, of which ||| would proceed to receive active therapy.13-15 In Canada, FL is common in people aged 50 years or older, and most are diagnosed at an advanced stage (i.e., 66% to 70% of patients are diagnosed at stage III or IV).23 FL can be further classified into 3 grades (1, 2, and 3 [a and b]) based on cell structures visible under the microscope, specifically the number of large FL cells (centroblasts).16 Grades 1, 2, and 3a diseases are generally considered low grade or slow growing compared to grade 3b disease, which grows fast and is considered high grade. FL may relapse or recur to more aggressive or high-grade forms of NHL, such as diffuse large B-cell lymphoma (DLBCL) during the disease, thus requiring other treatment options to manage the disease.3,24 In a UK study that prospectively followed 325 patients diagnosed with FL between 1972 and 1999, the risk of histologic conversion to more aggressive forms was reported to be 28% (95% CI, 23% to 34%) by 10 years of diagnosis.3,25 Some FL patients present with no symptoms, while others present with a range of symptoms, most of which are not specific to FL. Typical symptoms include adenopathy, splenomegaly, locally obstructing symptoms, fever, night sweats, and weight loss.1,3,26

Survival depends on the prognostic factors used in the FLIPI score.24 The FLIPI prognostic score system takes into account the following factors: age (> 60 years versus ≤ 60 years), Ann Arbor stage (III to IV versus I to II), number of involved nodal areas (> 4 versus ≤ 4), hemoglobin level (< 120 g/L versus ≥ 120 g/L), and serum lactate dehydrogenase concentration (above normal versus normal or below).27 Patients with “good” prognostic FLIPI scores respond well to treatment compared to patients with “poor” FLIPI scores, in whom cancer may recur following treatment.24 According to the Canadian Cancer Society, 91% of patients considered “low risk” at diagnosis according to the FLIPI score have a 5-year survival rate and 71% have a 10-year survival rate; 78% of patients considered “intermediate risk” have 5-year survival rate and 51% have a 10-year survival rate; and 53% of patients considered high risk have a 5-year survival rate and 36% have a 10-year survival rate.17 Casulo and colleagues (2015)28 reported that patients with POD24 after first-line rituximab therapy experienced particularly poor outcomes. In a previous study, the same authors reported that patients with POD24 had a 5-year survival rate of only 50% compared to 90% for patients without POD24.28

NHL, including FL, is diagnosed using immunohistochemical and genetic testing of tissue samples biopsied from lymph nodes.20 Patients undergo diagnostic testing to confirm r/r FL grade 1, 2, or 3a disease after 2 or more lines of systemic therapies. Testing may include a history and physical examination, tissue biopsy to confirm relapse or rule out transformation to aggressive lymphoma, imaging tests (PET scans or CT scans), and laboratory tests (e.g., complete blood counts). The timing of diagnostic tests relative to receiving axicabtagene ciloleucel may vary between patients.29

Standards of Therapy

Contents within this section have been informed by materials submitted by the sponsor and clinical expert input. The following have been summarized and validated by the CADTH review team.

The treatment goals for FL vary depending upon the stage of the FL and individual patient factors. In general, available treatments for stage I to II FL have curative potential; however, for most patients with stage III to IV FL, no curative therapies are available. Therefore, the main goals of treatment are to cure the lymphoma in patients with stage I to II FL, and to extend remission in patients with stage III to IV FL.30

Once a diagnosis of FL is confirmed, the gold standard for the management of asymptomatic patients with indolent FL is watchful waiting, also known as “watch and wait.”26,31 According to the clinical experts consulted by CADTH, watch and wait is a common practice for many patients with FL, even after disease relapse.

First-Line Treatments

For small, localized symptomatic FL, radiotherapy is considered the standard of care according to North American and European guidelines.30,32,33 This is supported by several provincial guidelines in Canada.34,35 For grade 1, 2, and 3a FL, the preferred chemoimmunotherapy regimen is bendamustine plus rituximab, based on high-level evidence of efficacy and favourable tolerability in this population.26,30,36 In frail and older patients, rituximab monotherapy, a chemotherapy-free approach, is the preferred first-line regimen according to European and North American guidelines.37,38 However, some treatment centres in Canada do not have access to rituximab monotherapy. Instead, physicians keep patients on bendamustine plus rituximab for several treatment cycles for as long as possible. Beyond first-line treatment, there is currently no gold standard of care for the r/r FL population.

Second-Line Treatments

Treatment options for second-line regimens for r/r FL depend on several factors, including level of fitness, prior treatment, and length of time to relapse.30 The preferred treatment strategy in this patient population is combined immunochemotherapy, such as O-CHOP (obinutuzumab, cyclophosphamide, doxorubicin hydrochloride [hydroxydaunorubicin], vincristine sulphate [Oncovin], and prednisone).30 SCT may be considered in young and fit patients with no comorbidities in the second-line setting. Auto-SCT is given more often than allo-SCT in this population. However, only a small subset of FL patients would be eligible for transplant in the second line. SCT is limited by highly selective eligibility criteria and is typically reserved for younger, medically fit patients with chemotherapy-sensitive disease.30,37,39,40 The clinical experts consulted by CADTH agreed with these strategies.

Third-Line Treatments

FL is a relapsing disease with continued unmet need in adult patients with r/r FL after 2 or more lines of therapy despite the availability of established therapies. Patients with r/r FL in the third- and later-line treatment setting represent a heavily pretreated and advanced-stage patient population. The standard of care in Canada for the third-line treatment of r/r FL is heterogenous and varies across regions. Based on Canadian clinician input, a heterogenous mix of immunochemotherapy (for most patients) and SCT (for a minority of patients) are the current treatment options in this hard-to-treat population.

Treatments in this setting may include SCT; however, there is controversy with regard to clinical benefit of SCT in patients with r/r FL, and both allo-SCT and auto-SCT may be associated with significant mortality and morbidity.30,37,39-41 While SCT has been included in the third-line treatment algorithm diagram for r/r FL, few people with r/r FL are expected to be eligible for SCT in the third line.

According to clinical experts consulted by the sponsor, most patients in the third-line treatment setting will continue recycling combined immunochemotherapy that might have been used in previous lines, including the following regimens (which are the most frequently used options in Canada): rituximab plus cyclophosphamide, vincristine, and prednisone (R-CVP); rituximab plus cyclophosphamide, hydroxydaunorubicin (doxorubicin hydrochloride), oncovin (vincristine sulphate), and prednisone (R-CHOP); obinutuzumab, cyclophosphamide, doxorubicin hydrochloride [hydroxydaunorubicin], oncovin (vincristine sulphate), and prednisone (O-CHOP); rituximab plus gemcitabine, dexamethasone, and cisplatin (R-GDP); bendamustine plus rituximab; and rituximab plus ifosfamide, carboplatin, and etoposide (R-ICE). Although it is used by some physicians in their clinical practice, lenalidomide plus rituximab regimen is currently not officially indicated for treatment of FL in Canada, as per the most recent Canadian product monograph for lenalidomide.42 Furthermore, although no official submission has been made by the sponsor to Canadian health technology assessment agencies, CADTH conducted a health technology review of lenalidomide plus rituximab in r/r B-cell NHL and concluded that available evidence remains limited.43 According to Canadian clinicians’ input, access to lenalidomide plus rituximab is somewhat limited in some provinces. In the third-line setting and beyond, idelalisib, a phosphoinositide 3-kinase (PI3K) inhibitor, is indicated for the treatment of patients with r/r FL in Canada.44 However, idelalisib is not publicly reimbursed across Canada and is inaccessible at some sites, according to several clinicians.45 Furthermore, it is generally at the bottom of the treatment list due to serious side effects and is, therefore, used as a palliative treatment. In recent years, CAR T-cell therapy has emerged as another form of immunotherapy for the treatment of blood cancers, including lymphomas. While promising results have been reported for the CAR T-cell therapies for advanced-stage lymphomas, severe adverse effects related to CAR T-cell therapy, such as CRS and neurologic toxicities, have also been reported.46 One CAR T-cell product, tisagenlecleucel, has been recently approved by Health Canada (December 2022) with a Notice of Compliance with conditions for the treatment of adult patients with r/r FL grade 1, 2, or 3a after 2 or more lines of systemic therapy.47 Note that at the time of writing this report, tisagenlecleucel has not been publicly funded for this indication.

In clinical practice, a patient’s response to treatment is commonly assessed using the Lugano classification,48 in which fluorodeoxyglucose-PET/CT is incorporated into the initial Ann Arbor Staging System for fluorodeoxyglucose-avid lymphomas.

Drug Under Review

Axicabtagene ciloleucel (Yescarta) is a CD19-directed genetically modified autologous T-cell immunotherapy (i.e., CAR T-cell therapy) that binds to CD19-expressing cancer cells and normal B cells.49 Following anti-CD19 CAR T-cell engagement with CD19-expressing target cells, the CD28 and CD3-zeta co-stimulatory domains activate downstream signalling cascades that lead to T-cell activation, proliferation, acquisition of effector functions, and secretion of inflammatory cytokines and chemokines. This sequence of events leads to killing of CD19-expressing cells.49 Axicabtagene ciloleucel is a single-dose, 1-time treatment in a patient-specific infusion bag. Axicabtagene ciloleucel should be administered by experienced health professionals in specialized treatment centres. Each patient-specific, single-infusion bag contains a suspension of anti-CD19 CAR-positive viable T cells in approximately 60 mL for a target dose of 2 × 106 anti-CD19 CAR T cells/kg body weight (range, 1 × 106 cells/kg to 2.4 × 106 cells/kg), to a maximum of 2 × 108 anti-CD19 CAR T cells for patients weighing 100 kg or more.49

Axicabtagene ciloleucel was approved in Canada on September 28, 2022, for the treatment of adult patients with r/r grade 1, 2, or 3a FL after 2 or more lines of systemic therapy. Axicabtagene ciloleucel has been previously reviewed by CADTH for 2 indications. On August 15, 2019, CADTH issued a positive recommendation for axicabtagene ciloleucel for adult patients with r/r large B-cell lymphoma after 2 or more lines of systemic therapy, including DLBCL not otherwise specified, primary mediastinal large B-cell lymphoma, high-grade B-cell lymphoma, and DLBCL arising from FL. On January 5, 2023, another positive recommendation was posted for the treatment of adult patients with DLBCL or high-grade B-cell lymphoma that is refractory to first-line chemoimmunotherapy or that relapses within 12 months of first-line chemoimmunotherapy, who are eligible for auto-SCT.

Key characteristics of axicabtagene ciloleucel are summarized in Table 3 with other treatments available for FL.

Table 3: Key Characteristics of Axicabtagene Ciloleucel

Characteristics

Axicabtagene ciloleucel49

Mechanism of action

A CD19-directed genetically modified autologous T-cell immunotherapy, that is, a CAR T-cell therapy that binds to CD19-expressing cancer cells and normal B cells

Indicationa

The treatment of adult patients with relapsed or refractory grade 1, 2, or 3a FL after 2 or more lines of systemic therapy

Route of administration

IV infusion for 1-time treatment

Recommended dose

1-time treatment

2 × 106 CAR-positive viable T cells/kg body weight (maximum of 2 × 108 cells/kg body weight)

Serious adverse effects or safety issues

  • CRS

  • Neurologic AEs

  • Prolonged cytopenias

  • Hypogammaglobulinemia

  • Serious infections

  • Secondary malignancies

  • Tumour lysis syndrome

AE = adverse event; CAR = chimeric antigen receptor; CRS = cytokine release syndrome; FL = follicular lymphoma.

aHealth Canada–approved indication.

Source: Yescarta Product Monograph.49

Stakeholder Perspectives

Patient Group Input

This section was prepared by the CADTH review team based on the input provided by patient groups. The full original patient input received by CADTH has been included in the Stakeholder section of this report.

One patient advocacy group, LC, provided input for this review. LC is a national Canadian registered charity whose mission is to empower patients and the lymphoma community through education, support, advocacy, and research. LC collaborates with patients, caregivers, health care professionals, other organizations, and stakeholders to promote early detection, find new and better treatments for lymphoma patients, help patients access treatments, learn about the causes of lymphoma, and work together to find a cure. The LC patient group expressed the need for accessible treatment options for patients, highlighting that local access to treatments will significantly improve patients' experience by reducing fear and the risk of getting sick while travelling and patient quality of life. The LC patient group expressed the need for accessible treatment options for patients, highlighting that local access to treatments will significantly improve patients' quality of life and experience by reducing fear and the risk of getting sick while travelling.

LC gathered information for this input via online surveys completed anonymously by patients between April 21, 2022, and April 3, 2023. Of the 143 responses submitted, 3 respondents reported having experience with axicabtagene ciloleucel. Of the total surveyed, 86% of respondents lived in Canada, 71% were aged between 55 and 64 years, 64% were female, and 34% had received a FL diagnosis within the last 3 to 5 years. Respondents indicated that fatigue (50%), body aches and pain (33%), enlarged lymph nodes (33%), indigestion (32%), and bodily swelling (21%) were the most challenging symptoms with the biggest impact on their quality of life at the time of diagnosis. Respondents described FL symptoms as challenges in their daily lives that impacted their ability to travel (46%), spend time with family or friends (41%), exercise (37%), concentrate (36%), and work or complete school or volunteer activities (35%). Anxiety or worry (84%), stress from diagnosis (77%), fear of progression (70%), and difficulty sleeping (48%) were the most common psychosocial symptoms that impacted patients.

About half (49%) of the respondents reported that they went through a period of watchful waiting before commencing treatment. Most respondents (43%) had received 1 line of treatment. The most common treatments reported by respondents who had received 1 or 2 lines of therapy included chemotherapy, chemoimmunotherapy, rituximab with or without bendamustine, or radiation. When asked to describe their treatment experience, 57% of respondents indicated that they were “satisfied” or “very satisfied” with the treatment options in the front-line setting. Only 22% of respondents indicated that they were “satisfied” or “very satisfied” with the current treatment options in the r/r setting. While describing how their current therapy (or most recent therapy) was able to manage their FL symptoms on a scale of 1 (strongly disagree) to 5 (strongly agree), 40% of respondents strongly agreed and 20% strongly disagreed. According to the respondents, the post-treatment symptoms that most significantly negatively impacted them included treatment-related fatigue (28%), immediate side effects of treatment (26%), and low activity level (23%). Fatigue (69%), hair loss (41%), and constipation (38%) were the most common side effects reported by respondents.

The most important outcomes (rated 5 on a scale of 1 to 5) included delaying disease progression (84%), longer disease remission (82%), improved quality of life, ability to perform daily activities (69%), ability to control disease symptoms (63%), and ability to normalize blood counts (58%). In total, 68% of respondents indicated a willingness to tolerate nonsevere side effects over the short-term period when undertaking a novel therapy, 42% expressed the importance of having a choice in deciding treatment options based on known side effects and expected outcomes, and 79% noted the need for more accessible treatment options that were proven to be effective for FL.

Two respondents completed all questions about axicabtagene ciloleucel in the survey. One survey respondent who confirmed that they had prior treatment experience with axicabtagene ciloleucel did not complete all other treatment questions. The 2 respondents who completed the survey reported having access to treatment via a clinical trial. One patient received treatment in the second-line and the other in the fifth-line setting. Reported side effects included CRS, neutropenia, febrile neutropenia, thrombocytopenia, constipation, and swelling. According to the respondents, the challenges that significantly impacted their physical and mental health during treatment included the frequent monitoring of side effects postinfusion, the inability to perform daily activities, and being away from family and friends. One respondent indicated they were away from home for 1 to 3 months, while the other was away for more than 3 months. Other highlighted challenges included financial issues due to absence from work and travel accommodation expenses accumulated during the clinical trial. Both respondents stated that they had a good or very good experience with axicabtagene ciloleucel and would recommend the treatment to other patients with r/r FL.

Clinician Input

Input From Clinical Experts Consulted by CADTH

All CADTH review teams include at least 1 clinical specialist with expertise regarding the diagnosis and management of the condition for which the drug is indicated. Clinical experts are a critical part of the review team and are involved in all phases of the review process (e.g., providing guidance on the development of the review protocol, assisting in the critical appraisal of clinical evidence, interpreting the clinical relevance of the results, and providing guidance on the potential place in therapy). In addition, as part of the review of axicabtagene ciloleucel, a panel of 4 clinical experts from across Canada was convened to characterize unmet therapeutic needs, assist in identifying and communicating situations where there are gaps in the evidence that could be addressed through the collection of additional data, promote the early identification of potential implementation challenges, gain further insight into the clinical management of patients living with a condition, and explore the potential place in therapy of the drug (e.g., potential reimbursement conditions). A summary of this panel discussion follows.

Unmet Needs

The clinical panel indicated that for patients with FL, the most important goals for an ideal treatment are to prolong survival (both OS and PFS) and to improve their quality of life. However, patients with r/r FL relapse after the front-line therapies or are refractory to the available treatments, which impacts their long-term PFS and quality of life. In addition, current treatments may not be well tolerated by some patients due to the related adverse events or complications associated with SCT. The clinical panel indicated that the greatest unmet needs for the current treatments exist for patients with cancer that progresses within 2 years after their initial therapy, for those who have already received auto-SCT or are ineligible for auto-SCT, and for those who have been double refractory to earlier line treatments (implying they have limited treatment options available to them). Patients who are eligible for CAR T-cell therapy but ineligible for auto-SCT would generally be older and less able to tolerate the auto-SCT process.

Place in Therapy

The treatment algorithm for adult patients with r/r FL is complicated. Many factors (e.g., patient characteristics, previous treatments, treatment effects and toxicity, drug plan coverage, disease progression and transformation, and patient preference) need to be considered before making decisions. Watch and wait is a common first-line approach for many patients with FL, and is also considered, though less commonly, after disease relapse. Patients who need active treatments typically receive bendamustine or rituximab-based therapies such as bendamustine plus rituximab, R-CVP, R-CHOP, or lenalidomide plus rituximab in the first- and second-line setting. PI3K inhibitors are rarely used. Patients with relapsed disease after treatment with chemoimmunotherapy, particularly those who progress within 2 years, may receive auto-SCT if they are suitable candidates. The clinical panel noted that some evidence suggests that auto-SCT cures half of the patients with POD24 and with PFS in the 20- to 30-year range. After all these treatments, some patients maintain the indolent status and some transform to large cell lymphoma; therefore, the proportion of patients who may be considered for treatment with CAR T-cell therapy is small. The clinical panel suggested that axicabtagene ciloleucel is most appropriate for use as a third or later line of treatment for patients with r/r FL. These patients usually have a treatment response that lasts less than 6 months from their last treatment (medication or SCT). There are not many options available for the patients at this stage. CAR T-cell therapy would be considered because it has a different mechanism of action.

The clinical panel noted that an auto-SCT is not mandatory before axicabtagene ciloleucel can be given, since this is not standard of care in Canada. The clinical panel suggested that an auto-SCT before axicabtagene ciloleucel would be recommended if the patient had access to auto-SCT and was eligible, noting that 80% of auto-SCT are performed after the first or second relapse.

For patients who have received previous CD19-targeted therapy, there is a lack of evidence to suggest whether the use of axicabtagene ciloleucel is appropriate.

Patient Population

The clinical panel indicated that, in practice, CAR T-cell therapy is used in a patient population that is broader than that in clinical trials, to which a more select population is recruited. For example, the panel noted that suitable candidates for treatment with axicabtagene ciloleucel would be patients with acceptable rather than excellent organ function, which is what is generally required for an auto-SCT. When determining whether axicabtagene ciloleucel treatment is suitable for a particular patient, bulk of disease and rapid disease progression are among the factors that need to be taken into account. In clinical practice, patients’ suitability can be determined based on clinical judgment, which combines medical history, laboratory and imaging findings, and often a lymph node biopsy.

The panel also noted that the patients who did not meet the eligibility criteria in the ZUMA-5 clinical trial (e.g., because they had certain comorbidities or disease status) would be the least suitable candidates for treatment with axicabtagene ciloleucel. Patients with an ECOG PS score of 3 or higher are also least suitable for treatment with axicabtagene ciloleucel.

The panel noted that there is no specific patient characteristic that can be used to predict who would respond better to axicabtagene ciloleucel than other patients.

Assessing the Response Treatment

The panel indicated that in clinical practice, patients are evaluated and followed in a manner similar to that described in the clinical trials of FL treatments. Remission and survival are measured. Physical exams and imaging exams are routinely conducted to assess the patient’s response to CAR T-cell therapy.

The panel suggested that meaningful responses to treatment with axicabtagene ciloleucel would include a high complete remission rate, durability of treatment response, and long-term PFS and OS. Ideally, a successful treatment would show a plateau in the PFS and OS curves. In addition, the clinicians were interested in knowing if the treatment is cost-effective. Ideally, the treatment benefits of axicabtagene ciloleucel can be compared to the other treatments.

The panel noted that after CAR T-cell therapy, clinicians would assess the treatment response (e.g., via CT scan) every 3 months, or sooner if needed.

When Patients Go Through Pretreatment but Do not Receive Axicabtagene Ciloleucel

The panel noted that situations when patients go through pretreatment but do not receive axicabtagene ciloleucel are rare. However, this can happen because of rapid disease progression in the interim or because the patient has major complications such as a new myocardial infarction or stroke. Manufacturing failure is another reason for this situation, although it is not expected to be an issue with axicabtagene ciloleucel.

If patients do not receive axicabtagene ciloleucel after undergoing pretreatment, most of them (in particular, high-risk patients) can progress within 6 months of their last treatment, and limited treatment options are then available for them. Palliative chemotherapy can be given. Other options may include radiation therapy, more chemotherapy, novel agents, or a clinical trial, depending on each patient’s clinical status.

Therapy Post Axicabtagene Ciloleucel Failure

The panel indicated that after infusion with axicabtagene ciloleucel and failure of treatment, patients may participate in a clinical trial. In the absence of a clinical trial, patients may try a different chemoimmunotherapy that they have not been exposed to or undergo auto-SCT if they have not already received this therapy.

Prescribing Considerations

The panel emphasized the importance of an accredited multidisciplinary team involving hematologists, infectious disease specialists, neurologists, an ICU team, and other specialists to diagnose, treat, and monitor the patients receiving axicabtagene ciloleucel and to ensure the safe and effective delivery of this treatment.

Clinician Group Input

This section was prepared by the CADTH review team based on the input provided by clinician groups. The full original clinician group input received by CADTH has been included in the Stakeholder section of this report.

Input from 1 clinician group, the OH-CCO Hematology Cancer Drug Advisory Committee, was summarized for this review. The OH-CCO Hematology Cancer Drug Advisory Committee provides timely, evidence-based clinical and health system guidance on drug-related issues supporting the OH-CCO's mandate, including the Provincial Drug Reimbursement Programs and the Systemic Treatment Program. Information in this input was obtained via video conferencing and email.

According to the clinicians consulted, the disease course of FL varies for every patient. Some patients may present with long remissions between therapies while others would have refractory disease. Current treatment goals for patients with FL, according to the clinician group, include palliative care and, in some scenarios, treatment with curative intent using allo-SCT. The most important goals outlined were to delay disease progression, improve patient HRQoL, and alleviate symptoms. Current standard of care for FL patients identified by the clinician group included chemotherapy, chemoimmunotherapy, auto-SCT, allo-SCT (for a selected group of patients), and radiation (to control symptoms and for palliative care scenarios). The OH-CCO Hematology Cancer Drug Advisory Committee acknowledged that current treatment options do not meet the needs of patients with r/r FL. The OH-CCO Hematology Cancer Drug Advisory Committee mentioned that patients who become refractory to chemotherapy have no other treatment options to delay the disease. In addition, the committee members highlighted that repeated administration of cytotoxic therapy may be associated with marrow damage (myelodysplastic syndrome), which further limits the ability to treat patients, and adversely affects quality of life. Hence, there is a need for treatment options that patients can tolerate. Treatment with a CAR T-cell therapy, according to the clinical group, is not anticipated to cause long-term marrow damage issues. The committee members said that a third-line therapy with a CAR T-cell therapy would be appropriate, given that current therapy provides less benefit to patients with r/r FL. The OH-CCO Hematology Cancer Drug Advisory Committee clinicians could not ascertain whether CAR T-cell therapy would replace auto-SCT; however, they said that they suspect that CAR T-cell therapy might be tried first, rather than auto-SCT, for patients with a history of chemotherapy-refractory forms of FL. The clinician group noted that there would be a prevalent FL patient population that will become eligible for axicabtagene ciloleucel at the time of its implementation. According to the clinicians, patients eligible to receive axicabtagene ciloleucel in clinical practice should be similar to patients included in the clinical trial. However, patients with severe organ dysfunction, poor performance status, and uncontrolled infections would be ineligible. The clinicians pointed out that patients who had received prior CD19-directed therapy (these patients were excluded from the trial) should be considered for treatment with CAR T-cell therapy and highlighted the need for flexibility around patients’ ECOG PS or KPS scores. The experts also noted that some patients might become ineligible to receive CAR T-cell therapy during the manufacturing process, which may lead to discontinuation. The clinician group noted that axicabtagene ciloleucel may have a toxicity profile that is different from that of tisagenlecleucel (Kymriah), another CAR T-cell therapy currently under review. The clinician group input aligned with the input provided by the clinical experts consulted during the CADTH review.

Drug Program Input

The drug programs provide input on each drug being reviewed through CADTH’s Reimbursement Review processes by identifying issues that may impact their ability to implement a recommendation. The implementation questions and corresponding responses from the clinical experts consulted by CADTH are summarized in Table 4.

Table 4: Summary of Drug Plan Input and Clinical Expert Response

Drug program implementation questions

Clinical expert response

Considerations for initiation of therapy

Should patients with the following be considered for treatment with axicabtagene ciloleucel?

  • ECOG PS > 1

  • prior CD19-targeted therapy (e.g., blinatumomab, tafasitamab)

  • prior allo-SCT

  • prior CAR T-cell therapy

  • active CNS involvement

  • other types of low-grade lymphoma (e.g., MZL, Waldenström macroglobulinemia, MALT lymphoma)

  • grade 3b FL

ECOG PS > 1: The clinical experts agreed that despite the ZUMA-5 trial being limited to patients with ECOG PS of 0 and 1, physicians would likely use axicabtagene ciloleucel in patients with ECOG PS of 2. Patients with an ECOG PS ≥ 3 would not be suitable candidates for treatment with axicabtagene ciloleucel.

Prior CD19-targeted therapy: The clinical experts had different opinions. Some suggested that any prior CD19-targeted therapy would preclude the use of axicabtagene ciloleucel. Others suggested that only patients who are refractory to CD19-targeted therapy (did not respond or relapsed within 6 months) would not be suitable candidates for treatment with axicabtagene ciloleucel.

Prior allogeneic transplant: The clinical experts had different opinions. Some suggested that prior allogeneic transplant would preclude the use of axicabtagene ciloleucel. Others suggested that axicabtagene ciloleucel should not be considered only if the allogeneic transplant was recent or if there were ongoing issues with graft-versus-host disease.

Prior CAR T-cell therapy: The clinical experts agreed that patients who have received prior CAR T-cell therapy should not be given axicabtagene ciloleucel.

Active CNS: The clinical experts agreed that patients with active CNS involvement should not be given axicabtagene ciloleucel.

Other types of low-grade lymphoma: The clinical experts noted that a small number of patients with MZL were included in the ZUMA-5 trial and axicabtagene ciloleucel would be expected to be efficacious in this population. There is a lack of evidence for Waldenström macroglobulinemia and MALT lymphoma. The clinical experts did not expect axicabtagene ciloleucel to be used in these populations.

Grade 3b FL: The clinical experts agreed that patients with grade 3b FL are not eligible for treatment with axicabtagene ciloleucel. The clinical experts noted that these patients would fall under the category of DLBCL and treatment decisions should be made from that perspective.

In the trial, monotherapy rituximab is not counted as a line of therapy. In some jurisdictions, single-agent rituximab is a funded option. What is the place in therapy for axicabtagene ciloleucel in these patients?

The clinical experts agreed with the design of the ZUMA-5 trial and did not believe that single-agent rituximab should be considered as a line of therapy. Single-agent rituximab is generally used for 4 weeks and then stopped. The clinical experts warned that considering the use of rituximab as a full line of therapy would make axicabtagene ciloleucel eligible earlier than is appropriate in the disease course.

Is there sufficient evidence to support re-treatment?

The clinical experts agreed that there was limited evidence to support re-treatment of patients with axicabtagene ciloleucel and that re-treatment would be unlikely to occur in the Canadian setting.

Considerations for prescribing of therapy

Delivery must take place at specialized treatment centres that are accredited and certified by the manufacturer.

There continues to be limited access to CAR T-cell therapy in Canada. While access is expanding, interprovincial travel or out-of-country funding remains necessary in many parts of Canada.

Due to geographical site limitations, patients may need to travel for treatment requiring interprovincial agreements to ensure equitable access.

Comment from the drug plans to inform pERC deliberations.

The provincial advisory group noted that tisagenlecleucel is also under review for r/r FL. Should the criteria for axicabtagene ciloleucel be aligned with that of tisagenlecleucel?

The clinical experts agreed that given the similarities between axicabtagene ciloleucel and tisagenlecleucel, the prescribing criteria should be aligned.

Generalizability

Should patients who recently started third or later line of systemic therapy be switched to CAR T-cell therapy provided all other criteria are met?

The clinical experts agreed that if a patient is responding to and tolerating a third or later line of therapy, it would not be appropriate to take them off that therapy and switch to axicabtagene ciloleucel.

Funding algorithm (oncology only)

Under what clinical circumstances would axicabtagene ciloleucel be used over tisagenlecleucel and vice-versa?

The clinical experts noted that they expect axicabtagene ciloleucel and tisagenlecleucel to differ in regard to safety profiles, specifically in terms of neurologic toxicity and CRS. A patient who may not be able to tolerate axicabtagene ciloleucel would be given tisagenlecleucel instead. No comparative evidence is available to inform this decision.

Care provision issues

Is postprogression biopsy needed to confirm that the disease has not transformed to DLBCL or other excluded histology before starting axicabtagene ciloleucel?

The clinical experts agreed that while a postprogression biopsy is preferred, it is not always feasible. As such, a postprogression biopsy should not be a requirement for access to axicabtagene ciloleucel.

System and economic issues

Feasibility of adoption (including budget impact) must be addressed. Although the sponsor estimates a low uptake for axicabtagene ciloleucel, the provincial advisory group is concerned that this may be an underestimate and that existing capacity may not be able to meet demand.

Comment from the drug plans to inform pERC deliberations.

If manufacturing delays occur, how would this impact the clinical effectiveness of axicabtagene ciloleucel?

The clinical experts noted that given the slow growing nature of r/r FL, manufacturing delays are not expected to significantly impact clinical effectiveness like might occur with other, faster growing cancers.

CAR = chimeric antigen receptor; CNS = central nervous system; CRS = cytokine release syndrome; DLBCL = diffuse large B-cell lymphoma; ECOG PS = Eastern Cooperative Oncology Group Performance Status; FL = follicular lymphoma; MALT = mucosa-associated lymphoid tissue; MZL = marginal zone lymphoma; pERC = CADTH pan-Canadian Oncology Drug Review Expert Review Committee; r/r = relapsed or refractory.

Clinical Evidence

The objective of this Clinical Review was to review and critically appraise the clinical evidence submitted by the sponsor on the beneficial and harmful effects of axicabtagene ciloleucel, target dose of 2 × 106 CAR T cells/kg body weight, to a maximum of 2 × 108 CAR-positive viable T cells, by IV infusion, in the treatment of adult patients with r/r FL after 2 or more lines of systemic therapy. The focus is to compare axicabtagene ciloleucel to relevant comparators and identify gaps in the current evidence.

A summary of the clinical evidence included by the sponsor in the review of axicabtagene ciloleucel is presented in 2 sections, with CADTH’s critical appraisal of the evidence included at the end of each section. The first section, the systematic review, includes a pivotal study that was selected according to the sponsor’s systematic review protocol. The second section includes 1 additional study that was considered by the sponsor to address important gaps in the systematic review evidence.

Included Studies

Clinical evidence included in the CADTH review and appraised in this document:

Systematic Review

Contents within this section have been informed by materials submitted by the sponsor. The following have been summarized and validated by the CADTH review team.

Description of Studies

Details of the included studies are summarized in Table 5.

Table 5: Details of Studies Included in the Systematic Review

Detail

ZUMA-5

Designs and populations

Study design

Open-label, single-arm, interventional phase II

Locations

17 sites in 2 countries: 15 sites in the US and 2 sites in France

Patient enrolment dates

||||| ||||| |||| |||||||| ||||| |||| ||||

Randomized (N)

N = 127 (FL, enrolled); N = 25 (MZL, enrolled)

  • The ZUMA-5 trial evaluated the efficacy of axicabtagene ciloleucel in r/r iNHL of FL and MZL histological subtypes. The indication to be reviewed is for FL only, which is the focus of the summary presented here.

n = 124 (received axicabtagene ciloleucel; safety analysis set)

n = 86 (inferential analysis set)

Inclusion criteria

  • Eligible patients were aged 18 years or older and had histologically confirmed diagnosis of B-cell iNHL, with histological subtype limited to FL (grade 1, grade 2, or grade 3a).

  • Eligible patients had r/r disease after 2 or more lines of therapy (which must have included an anti-CD20 monoclonal antibody combined with an alkylating agent; single-agent anti-CD20 antibody did not count as an eligible line of therapy), and at least 2 weeks or 5 half-lives must have elapsed since any prior systemic therapy. Patients with stable disease (without relapse) > 1 year from completion of last therapy were not eligible.

  • Eligible patients had at least 1 measurable lesion according to the Lugano Response Criteria for Malignant Lymphoma48 and an ECOG PS score of 0 or 1.

Exclusion criteria

  • Patients were excluded if they had transformed FL, FL histological grade 3b, small lymphocytic lymphoma, lymphoplasmacytic lymphoma, or a history of malignancy other than nonmelanoma skin cancer or carcinoma.

  • Patients were excluded if they had prior allo-SCT, auto-SCT within 6 weeks of planned axicabtagene ciloleucel infusion, CD19-targeted therapy, or CAR or other genetically modified T-cell therapies.

  • Patients were ineligible if they had an infection or a history of CNS disorders, CSF malignant cells, cardiovascular disease, autoimmune disease, DVT or pulmonary embolism, or severe immediate hypersensitivity reaction to any of the agents used in this study.

Drugs

Intervention

Patients received conditioning chemotherapy of fludarabine (30 mg/m2/day) and cyclophosphamide (500 mg/m2/day) administered over 3 days, 3 to 5 days before axicabtagene ciloleucel infusion.

Axicabtagene ciloleucel infusion (target dose of 2 × 106 anti-CD19 CAR T cells/kg body weight).

Study duration

Screening phase

28 days

Conditioning phase

Day –5 to day –3

Treatment phase

Day 0 single day infusion

Follow-up phase

Every 3 months (from month 6 to month 18)

Every 6 months (from month 24 to month 60)

Every 12 months (from month 72 to year 15)

Outcomes

Primary end point

ORR by central assessment (18 and 36 months)

Secondary and exploratory end points

Secondary (18 and 36 months):

  • CRR by central assessment

  • ORR and CRR by central assessment in patients with ≥ 3 lines of prior therapy

  • ORR by investigator assessment

  • Best overall response (CR, PR, stable disease, progressive disease, or not evaluable as best response to treatment) by central and investigator assessment

  • DOR by central and investigator assessment

  • PFS by central and investigator assessment

  • OS

  • TTNT

  • Incidence of TEAEs and clinically significant changes in laboratory values

  • Incidence of immunogenicity against the anti-CD19 CAR

  • Levels of anti-CD19 CAR T cells in blood

  • Levels of cytokines in serum

Publication status

Publications

Jacobson et al. (2020)50

Jacobson et al. (2020)51

Neelapu et al. (2021)52

Jacobson et al. (2022)53

Clinical trial number

NCT0310533618

allo-SCT = allogeneic stem cell transplant; auto-SCT = autologous stem cell transplant; CAR = chimeric antigen receptor; CNS = central nervous system; CR = complete response; CRR = complete response rate; CSF = cerebrospinal fluid; DOR = duration of response; DVT = deep vein thrombosis; ECOG PS = Eastern Cooperative Oncology Group Performance Status; FL = follicular lymphoma; iNHL = indolent non-Hodgkin lymphoma; MZL = marginal zone lymphoma; ORR = objective response rate; OS = overall survival; ORR = objective response rate; PFS = progression-free survival; PR = partial response; r/r = relapsed or refractory; TEAE = treatment-emergent adverse event; TTNT = time to next treatment.

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: ZUMA-5 Clinical Study Report.18

The ZUMA-5 trial is a multicentre, international, open-label, single-arm phase II trial. The study objective was to determine the efficacy and safety of axicabtagene ciloleucel in patients with r/r FL or MZL after 2 or more lines of systemic therapy. In line with the submitted reimbursement request and anticipated Health Canada indication, the r/r FL patient group will be the focus of this review. The ZUMA-5 study design, shown in Figure 1, consisted of screening, enrolment/leukapheresis, a conditioning chemotherapy period, a 1-time IV infusion of axicabtagene ciloleucel, posttreatment assessment, and long-term follow-up.

Between |||| |||| ||| |||| ||||, 127 FL patients were enrolled at 15 sites in the US and 2 in France. There were no study sites in Canada. Prior to receiving any treatments, patients underwent leukapheresis to obtain T cells as part of the manufacturing process for axicabtagene ciloleucel. Patients were then treated with cyclophosphamide and fludarabine lymphodepleting chemotherapy between 5 and 3 days before axicabtagene ciloleucel infusion. After 2 days of rest, patients received axicabtagene ciloleucel through IV infusion at a target dose of 2 × 106 anti-CD19 CAR T cells/kg body weight. PET-CT scans for disease assessment were conducted at week 4, month 3, month 6, month 9, month 12, month 15, month 18, month 24, and at any subsequent scheduled or unscheduled visit if disease progression was a clinical concern, as per standard of care.

Analyses were conducted at 18 months, 24 months (not presented), and 36 months. The statistical analysis plan prespecified that tests be conducted on the inferential analysis set at 18 months; this was defined as the point when 80 patients had been followed for at least 18 months. Using all enrolled patients, analyses were conducted at 18 months, 24 months, and 36 months; this population of patients is referred to as the FAS. The data cut-off for the 18-month analysis was September 14, 2020, and the data cut-off for the 36-month analysis was March 31, 2022.

Figure 1: ZUMA-5 Study Design

This figure shows the schema of the ZUMA-5 clinical trial, beginning with screening, and followed by, in this order, enrolment/leukapheresis, the conditioning chemotherapy period, the investigational product treatment period, the posttreatment assessment period, and finally, the long-term follow-up period.

CAR = chimeric antigen receptor.

Source: Reproduced as is from ZUMA-5 Clinical Study Report.18

Populations

Inclusion and Exclusion Criteria

Patients eligible for inclusion in the ZUMA-5 trial were aged 18 years or older, with a confirmed diagnosis of r/r indolent NHL, with FL limited to grade 1, 2, or 3a, and with r/r disease after 2 or more lines of systemic therapy. Prior therapy must have included an anti-CD20 monoclonal antibody combined with an alkylating agent. Eligible patients were required to have least 1 measurable lesion according to the Lugano Response Criteria for Malignant Lymphoma,48 an ECOG PS score of 0 or 1, and no known history of central nervous system involvement. Patients were excluded from the ZUMA-5 trial if they had transformed disease or if they had received auto-SCT within 6 weeks of the planned axicabtagene ciloleucel infusion.

Interventions

Five days before axicabtagene ciloleucel infusion, patients received an IV conditioning chemotherapy regimen of fludarabine 30 mg/m2/day and cyclophosphamide 500 mg/m2/day. Patients were treated with these chemotherapies over 3 days to induce lymphocyte depletion. Following 2 days of rest, patients received axicabtagene ciloleucel infusion at a target dose of 2 × 106 anti-CD19 CAR T cells/kg body weight. Patients who achieved a partial response at the 3-month assessment but subsequently experienced disease progression were eligible for an optional course of re-treatment with conditioning chemotherapy and axicabtagene ciloleucel.

Corticosteroid therapy at a pharmacologic dose (≥ 5 mg/day of prednisone or equivalent doses of other corticosteroids) and other immunosuppressive drugs were restricted for 7 days before leukapheresis and 5 days before axicabtagene ciloleucel administration. Systemic corticosteroids were restricted as premedication to patients for whom CT scans with contrast were contraindicated (i.e., patients with contrast allergy or impaired renal clearance). Corticosteroids and other immunosuppressive drugs were restricted for 3 months after axicabtagene ciloleucel administration unless used to manage axicabtagene ciloleucel–related or other severe toxicities (e.g., anaphylaxis). Treatments for the patient’s lymphoma other than what was defined/allowed in the protocol, such as chemotherapy, immunotherapy, targeted agents, radiation, high-dose corticosteroid, and other investigational agents, were prohibited, except as needed for treatment of disease progression after axicabtagene ciloleucel infusion.

Outcomes

A list of the efficacy end points assessed in this review is provided in Table 6. Summarized end points are based on outcomes included in the sponsor’s Summary of Clinical Evidence as well as any outcomes identified as important according to the clinical experts consulted by CADTH and stakeholder input from patient and clinician groups and public drug plans. Using the same considerations, the CADTH review team selected end points they considered to be most relevant for informing the CADTH Canadian Drug Expert Committee (CDEC) in its deliberations and finalized this list of end points in consultation with members of the expert committee.

The primary end point of the ZUMA-5 trial was ORR, defined as the incidence of a complete response (CR) or a partial response, as determined by central assessment. These end points were defined by the Lugano classification criteria. Key secondary end points determined by central assessment included the CRR, defined as the incidence of CR as the best response to treatment, and the ORR and CRR in patients who had 3 or more lines of prior therapy. Other secondary end points included the best overall response, defined as the incidence of CR, partial response, stable disease, progressive disease, or NE as the best response to treatment, as adjudicated by central assessment. The DOR was measured in patients who had an objective response and was defined as the time from the first objective response to disease progression or death. PFS was defined as the time from the date of axicabtagene ciloleucel infusion for the inferential or safety analysis sets (or date of leukapheresis for the FAS) to the date of disease progression or death, while OS was defined as the time from the date of axicabtagene ciloleucel infusion for the inferential or safety analysis sets (or date of leukapheresis for the FAS) to the date of death. TTNT was defined as the time from the date of axicabtagene ciloleucel infusion to the start of new lymphoma therapy or death. Patient-reported outcomes were not reported in the ZUMA-5 trial.

Safety outcomes that occurred with the onset or after infusion of axicabtagene ciloleucel were reported as TEAEs. The incidence of TEAEs and SAEs were summarized by preferred term, according to the Medical Dictionary for Regulatory Activities (MedDRA) Version 25.0. The severity of AEs was graded using the National Cancer Institute’s Common Terminology Criteria for Adverse Events Version 4.03 or more recent editions. AEs of special interest were also reported and include CRS, neurologic events, cytopenias, infection, and hypogammaglobulinemia.

Table 6: Outcomes Summarized From the Studies Included in the Systematic Review

Outcome measure

Time point

ZUMA-5

ORR by central assessment

18 months

Primary

CRR by central assessment

18 months

Secondary

ORR and CRR by central assessment in patients with ≥ 3 lines of prior therapya

18 months

Secondary

Best overall response (CR, PR, stable disease, progressive disease, or not evaluable as best response to treatment) by central and investigator assessment

18 months

Secondary

ORR by investigator assessment

18 and 36 months

Secondary

DOR by central assessment

18 months

Secondary

DOR by investigator assessment

18 and 36 months

Secondary

PFS by central assessment

18 months

Secondary

PFS by investigator assessment

18 and 36 months

Secondary

OS

18 and 36 months

Secondary

TTNT

18 and 36 months

Secondary

Safety outcomes after infusion of axicabtagene ciloleucel

18 and 36 months

Secondary

CR = complete response; CRR = complete response rate; DOR = duration of response; ORR = objective response rate; OS = overall survival; PFS = progression-free survival; PR = partial response; TTNT = time to next treatment.

aAll other outcomes were analyzed in patients with 2 or more lines of prior therapy unless otherwise specified.

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: ZUMA-5 Clinical Study Report.18

Statistical Analysis

The FAS of the ZUMA-5 trial consists of all enrolled patients (n = 127). The inferential analysis set was defined as the first 80 patients followed for 18 months after axicabtagene ciloleucel treatment. For any patient who could have but did not attend the 18-month study visit, an additional patient was added, resulting in 86 patients included in the inferential analysis set. This sample size (n = 86) provided 93% power to reject the null hypothesis of an ORR of less than or equal to 40% at the alpha level of 0.0237 under an assumed alternative ORR of 60%.

An interim analysis was conducted for efficacy outcomes when 30 patients had been followed for 6 months after axicabtagene ciloleucel infusion. The nominal alpha level for the assessment of efficacy for this analysis was 0.0003. Another interim analysis was conducted when 80 patients had been followed for 6 months after axicabtagene ciloleucel infusion. The nominal alpha level for the assessment of efficacy for this analysis was 0.0005. Another interim analysis was conducted when at least 80 patients had been followed for at least 9 months after the first disease response assessment. The nominal alpha level for the assessment of efficacy for this analysis was 0.0005. Primary analysis was to be performed when at least 80 patients in the inferential analysis set had been followed for 12 months after the first disease response assessment, reserving alpha of 0.0237 for the final significance test.

At each interim analysis, 4 hypotheses were tested in the inferential analysis set using a fixed sequence procedure to control overall type I error at 1-sided alpha level of 0.025. In order, the hypotheses tested were ORR as determined by central assessment, CRR as determined by central assessment, ORR as determined by central assessment in the patients who had had 3 or more lines of prior therapy, and CRR as determined by central assessment in the patients who had had 3 or more lines of prior therapy. Each significance test used the alpha allocated at the time of interim analysis.

Analyses conducted on the FAS at the 18-month and 36-month cut-off dates were intended to be descriptive, and power calculations were not conducted.

Subgroup Analyses

Prespecified baseline subgroups used to examine key efficacy and safety analyses include age (< 65 years, ≥ 65 years), sex, race, ethnicity, FLIPI total score, ECOG PS score (0, 1), meeting the criteria for high tumour bulk load as per the Groupe d’Étude des Lymphomes Folliculaires (GELF) versus not meeting these criteria, relapsed versus refractory at study entry, time to relapse from initiation of first anti-CD20 chemotherapy combination therapy (≥ 24 months, < 24 months), prior treatment with PI3K inhibitor, number of lines of prior therapy, and double refractory status.

Table 7: Statistical Analysis of Efficacy End Points

End point

Statistical model

Adjustment factors

Handling of missing data

Sensitivity analyses

ORRa

95% CI calculated using the Clopper-Pearson method

NA

Patients who do not meet the criteria for an ORR by the analysis cut-off date will be considered nonresponders.

Conducted in the FAS and the safety analysis set

Best overall response rate (including CRR)a

95% CI calculated using the Clopper-Pearson method

NA

Derivation of this end point only includes response assessments obtained after initiation of axicabtagene ciloleucel infusion and up to progressed disease or the disease assessments before subsequent anticancer therapy.

Conducted in the FAS and the safety analysis set

DOR

KM estimates, 2-sided 95% CI

NA

Patients who do not meet the criteria for progression or death by the analysis cut-off date will be censored at their last evaluable disease assessment date.

Conducted in the FAS and the safety analysis seta

PFS

KM estimates, and 2-sided 95% CI

NA

Patients who do not meet the criteria for progression by the analysis cut-off date will be censored at their last evaluable disease assessment date.

Conducted in the FAS and the safety analysis seta

OS

KM estimates, 2-sided 95% CI

NA

Patients who have not died by the analysis cut-off date will be censored by their last date known to be alive before the data cut-off date.

Conducted in the FAS and the safety analysis set

TTNT

KM estimates, 2-sided 95% CI

NA

Patients who have not received subsequent new therapy and are still alive will be censored at the last contact date.

Conducted in the FAS and the safety analysis set

CI = confidence interval; CR = complete response; DOR = duration of response; FAS = full analysis set; KM = Kaplan-Meier; NA = not applicable; ORR = objective response rate; OS = overall survival; PFS = progression-free survival; TTNT = time to next treatment.

aPer central assessment.

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: ZUMA-5 Clinical Study Report.18

Analysis Populations

A summary of the analysis populations in the ZUMA-5 trial is shown in Table 8.

Results

Patient Disposition

A summary of patient disposition in the ZUMA-5 trial is presented in Table 9. At the 36-month analysis, ||| patients with either FL or MZL had been screened, with 127 patients enrolled in the FL group and included in the FAS. Discontinuation from the study occurred for ||| of patients, with the majority discontinuing due to death (|||||). Median follow-up at the 36-month analysis was ||||| |||||||||||||| || ||| || |||| ||||||). The safety analysis set included only the 124 patients who received an axicabtagene ciloleucel infusion. Patient disposition for the inferential 18-month analysis was not reported.

Table 8: Analysis Populations in the ZUMA-5 Trial

Population

Definition

FAS

All enrolled (leukapharesed) patients

Safety analysis set

All patients treated with any dose of axicabtagene ciloleucel

Inferential analysis set

Enrolled patients treated with any dose of axicabtagene ciloleucel who met the eligibility criteria stipulated in protocol amendment 2 or higher.

The first 86 patients treated with axicabtagene ciloleucel who reached the 18-month follow-up were included.

Re-treatment analysis set

All participants who underwent re-treatment with axicabtagene ciloleucel

FAS = full analysis set.

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: ZUMA-5 Clinical Study Report.18

Table 9: Summary of Patient Disposition From Studies Included in the Systematic Review

Patient disposition

ZUMA-5

Axicabtagene ciloleucel

FASa

18 months

(N = 127)

Axicabtagene ciloleucel

FASa

36 months

(N = 127)

Screened, N

181b

||||

Reason for screening failure, n (%)

|| ||||

|| ||||

    Failed to meet eligibility criteria, n

||

||

    Investigator decision, n

||

||

Enrolled, N (%)

127

127

Discontinued from study, n (%)

|| ||||

|| ||||

Reason for discontinuation by patients who did not receive axicabtagene ciloleucel, n (%)

| |||

| |||

    Death

| |||

| |||

    Patient withdrawal of consent from further follow-up

||

||

    Other

| |||

| |||

Reason for discontinuation by patients who received axicabtagene ciloleucel, n (%)

|| ||||

|| ||||

    Death

|| ||||

|| ||||

    Investigator decision

| |||

| |||

    Lost to follow-up

| |||

| |||

    Patient withdrawal of consent from further follow-up

| |||

| |||

FAS, N

127

127

    Follow-up time (months), mean (SD)

||||| ||||||

||||| |||||||

    Follow-up time (months), median (range)

||||| |||| || |||||

||||| |||| || |||||

PP, N

124

124

Safety, N

124

124

FAS = full analysis set; PP = per protocol; SD = standard deviation.

aPatient disposition was not reported for the 18-month inferential analysis set.

bReported number of patients screened includes patients with follicular lymphoma (FL) or marginal zone lymphoma. The number of patients with FL only was not reported.

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: ZUMA-5 Clinical Study Report.18

Baseline Characteristics

A summary of baseline characteristics in the ZUMA-5 trial is shown in Table 10. In the FAS, the median age was 60 years (range, 34 years to 79 years) and 62% of patients had an ECOG PS score of 0. Of the enrolled patients, 69% were refractory, defined as progressing within 6 months of their most recent treatment. Most patients enrolled in the ZUMA-5 trial had received 2 prior therapies (|||), 26% had received 3 prior therapies, 20% had received 4 prior therapies, and 17% had received 5 or more prior therapies. The proportion of patients who had received a prior auto-SCT was 24%, while the proportion of patients with high bulk tumour was 51%. The proportion of patients who had progressed within 24 months of anti-CD20 chemotherapy combination therapy (i.e., POD24) was 55%.

The baseline characteristics outlined in Table 10 are limited to those most relevant to this review as prognostic or effect-modifying variables.

Table 10: Summary of Baseline Characteristics From Studies Included in the Systematic Review

Characteristic

ZUMA-5

Inferential analysis set

18 months

(N = 86)

FAS

36 months

(N = 127)

Age (years), median (range)

|| ||| || |||

60 (34 to 79)

  < 65 years, n (%)

|| ||||

87 (69)

Male, n (%)

48 (56)

75 (59)

Female, n (%)

38 (44)

52 (41)

Race, n (%)

  Asian

| |||

| |||

  Black or African American

| |||

| |||

  White

|| ||||

||| ||||

  Other

| |||

| |||

ECOG PS, n (%)

  0

|| ||||

79 (62)

  1

|| ||||

48 (38)

Histological category at study entry, n (%)

  Grade 1

20 (23)

34 (27)

  Grade 2

43 (50)

63 (50)

  Grade 3a

23 (27)

30 (24)

Disease stage, n (%)

  I

2 (2)

| |||

  II

9 (10)

|| ||||

  III

35 (41)

|| ||||

  IV

40 (47)

|| ||||

FLIPI total score, n (%)

  0

3 (3)

| |||

  1

10 (12)

|| ||||

  2

33 (38)

|| ||||

  3

25 (29)

|| ||||

  4

12 (14)

|| ||||

  5

3 (3)

| |||

  Low risk (0 to 1)

13 (15)

|| ||||

  Intermediate risk (2)

33 (38)

|| ||||

  High risk (3 to 5)

40 (47)

56 (44)

Relapsed or refractory subgroupa

  Relapsed

23 (27)

40 (31)

  Refractory

63 (73)

87 (69)

Number of prior therapies, n (%)

  1

| |||

| |||

  2

|| ||||

|| ||||

  3

|| ||||

33 (26)

  4

|| ||||

25 (20)

  ≥ 5

|| ||||

22 (17)

Response to last line of therapy, n (%)

  CR

|| ||||

|| ||||

  PR

|| ||||

|| ||||

  Stable disease

|| ||||

|| ||||

  Progressive disease

|| ||||

|| ||||

  NE

| |||

| |||

  Unknown

| ||||

|| ||||

Receiving prior auto-SCT

21 (24)

30 (24)

POD24, n (%)

49 (57)

70 (55)

High tumour bulk, n (%)

|| ||||

65 (51)

Prior therapies, n (%)

  PI3K inhibitor

|| ||||

36 (28)

  Anti-CD20 single agent

|| ||||

40 (31)

  Alkylating agent

|| ||||

|| ||||

  Anti-CD20 + alkylating agent

|| |||||

||| ||||

  Lenalidomide

|| ||||

38 (30)

auto-SCT = autologous stem cell transplant; CR = complete response; ECOG PS = Eastern Cooperative Oncology Group Performance Status; FAS = full analysis set; FLIPI = Follicular Lymphoma International Prognostic Index; NE = not evaluable; PI3K = phosphoinositide 3-kinase; POD24 = progression of disease within 24 months; PR = partial response.

aPatients with FL who progressed within 6 months of completion of the most recent prior treatment are defined as refractory. Patients with FL who progressed 6 months after completing the most recent prior treatment are defined as relapsed.

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: ZUMA-5 Clinical Study Report.18

Exposure to Study Treatments

As axicabtagene ciloleucel is a single infusion, treatment exposure measures such as total patient-weeks, duration, and adherence are not applicable. Patients in the ZUMA-5 trial were eligible for re-treatment (n = 11 in the 18-month FAS and n = 13 in the 36-month FAS). It is not anticipated that axicabtagene ciloleucel will be eligible for use in re-treatment in Canada.

Table 11: Summary of Concomitant and Subsequent Treatment in the ZUMA-5 Trial

Exposure

ZUMA-5

Safety analysis set

18 months

(N = 124)

Safety analysis set

36 months

(N = 124)

Concomitant medications and procedures

Received concomitant therapy of interest, n (%)

|| ||||

|| ||||

Corticosteroids, n (%)

|| ||||

|| ||||

Tocilizumab, n (%)

|| ||||

|| ||||

Vasopressors

|| ||||

|| ||||

Nonsteroidal immunosuppressive agents

| |||

| |||

Immunoglobulins

|| ||||

|| ||||

Endotracheal intubation

| |||

| |||

Subsequent therapy

  Received subsequent stem cell transplant, n (%)

| |||

| |||

  Received subsequent anticancer therapy, n (%)

|| ||||

|| ||||

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: ZUMA-5 Clinical Study Report.18

Efficacy

Overall Survival

A summary of the OS results from the ZUMA-5 trial is shown in Table 12. At the 36-month FAS time point, ||| of patients had died due to any cause, and the median OS had not been reached. The KM-estimated survival probability at 18 months was ||||| ||||| || ||||||, at 24 months was ||||| ||||| || |||||, and at 36 months was 75.5% (95% CI, 66.9% to 82.2%). Results for the 18-month inferential analysis set and FAS are shown for context. The estimated OS curve in the ZUMA-5 trial is shown in Figure 2.

Table 12: OS in the ZUMA-5 Trial

Characteristic

ZUMA-5

Inferential analysis set

18 months

(N = 86)

FAS

18 months

(N = 127)

FAS

36 months

(N = 127)

Death from any cause, n (%)

|| ||||

|| ||||

|| ||||

Alive, n (%)

|| ||||

||| ||||

|| ||||

OS time (months), median (95% CI)

NE (31.6 to NE)

|| |||| |||

NE (NE to NE)

  Survival probability at 18 months,% (95% CI)a

88.3 (79.4 to 93.5)

|||| |||||| |||||

|||| |||||||||||

  Survival probability at 24 months, % (95% CI)a

|||| |||||||||||

|||| |||||||||||

|||| |||||||||||

  Survival probability at 36 months, % (95% CI)a

NE

||

75.5 (66.9 to 82.2)

CI = confidence interval; FAS = full analysis set; NE = not evaluable; OS = overall survival.

aSurvival probabilities are according to Kaplan-Meier estimates.

Source: ZUMA-5 Clinical Study Report.18

Figure 2: Redacted

This figure has been redacted.

CI = confidence interval; FL = follicular lymphoma; NE = not evaluable.

This figure has been redacted.

Source: ZUMA-5 Clinical Study Report.18

Progression-Free Survival

A summary of the PFS results from the ZUMA-5 trial is shown in Table 13. At the 36-month FAS time point, ||| of patients had experienced a progression event. The median PFS was 40.2 months (95% CI, 28.9 months to NE). The KM-estimated PFS rate at 18 months was ||||| ||||| || |||||, at 24 months was ||||| ||||| || |||||, and at 36 months was 54.4% (95% CI, 44.2% to 63.5%). Results for the 18-month inferential analysis set and 18-month FAS are shown for context. The estimated PFS curve in the ZUMA-5 trial is shown in Figure 3.

Table 13: PFS in the ZUMA-5 Trial

Characteristic

ZUMA-5

Inferential analysis set

18 months

(N = 86)

FAS

18 months

(N = 127)

FAS

36 months

(N = 127)

Events, n (%)

|| ||||

|| ||||

|| ||||

Censored, n (%)

|| ||||

|| ||||

|| ||||

PFS time (months), median (95% CI)

NE (23.5 to NE)

|| |||| |||

40.2 (28.9 to NE)

  PFS probability at 18 months, % (95% CI)a

68.8 (57.4 to 77.8)

|||| |||||||||||

|||| |||||||||||

  PFS probability at 24 months, % (95% CI)a

|||| |||||||||||

|||| |||||||||||

|||| |||||||||||

  PFS probability at 36 months, % (95% CI)a

NE

||

54.4 (44.2 to 63.5)

CI = confidence interval; FAS = full analysis set; NE = not evaluable; PFS = progression-free survival.

Note: PFS is reported according to central assessment at the 18-month analysis and as per investigator assessment for the 36-month analysis.

aSurvival probabilities are according to Kaplan-Meier estimates.

Source: ZUMA-5 Clinical Study Report.18

Figure 3: Redacted

This figure has been redacted.

CI = confidence interval; FAS = full analysis set; NE = not evaluable; PFS = progression-free survival.

This figure has been redacted.

Note: PFS is reported as per investigator assessment for the 36-month analysis.

Source: ZUMA-5 Clinical Study Report.18

Objective Response Rate

A summary of the response results for the ZUMA-5 trial is shown in Table 14. At the 36-month FAS time point, the ORR as per investigator assessment was 94% (95% CI, 88% to 97%), while the CRR was 79% (|| || ||). The primary end point in the ZUMA-5 trial was ORR at the 18-month analysis in the inferential analysis set. The estimated ORR as per central assessment was 94% (|| || ||) and the CRR was 79% (|| || ||), sufficient to reject the null hypotheses of 40% for ORR and 15% for CRR.

Table 14: ORR in the ZUMA-5 Trial

Characteristic

ZUMA-5

Inferential analysis set

18 months

(N = 86)

FAS

18 months

(N = 127)

FAS

36 months

(N = 127)

ORR, n (%, [95% CI]a)

81 (94 [|| || ||])

||| ||| ||| || ||||

119 (94 [88 to 97])

    P value

NAb

NA

NA

CRR, n (%, [95% CI]a)

68 (79 [||| || ||])

|| ||| ||| || ||||

100 (79 [|| || ||])

    P value

NAb

NA

NA

PR, n (%)

13 (15)

|| ||||

19 (15)

Stable disease, n (%)

| |||

| |||

2 (2)

Progressive disease, n (%)

| |||

| |||

2 (2)

Undefined/no disease, n (%)

| |||

| |||

||

Not done, n (%)c

| |||

| |||

4 (3)

CI = confidence interval; CRR = complete response rate; FAS = full analysis set; NA = not applicable; NE = not evaluable; ORR = objective response rate; PR = partial response.

a95% CI from the Clopper-Pearson method.

bHypothesis testing was conducted on an earlier 18-month data cut-off with 84 patients yielding significant P values (P < 0.0001) for both ORR and CRR.

cPatients who were treated with axicabtagene ciloleucel but died before first disease assessment were recorded as having a “not done” response.

Note: ORR is reported according to central assessment at the 18-month analysis and as per investigator assessment for the 36-month analysis.

Source: ZUMA-5 Clinical Study Report.18

The ORR results for selected subgroups, based on discussion with clinical experts consulted by CADTH, are presented in Table 15. The results appear to be consistent across all subgroups, with the ORR from |||| ||| || |||| in patients who received 1 prior line of therapy to ||| ||| || ||| in patients whose disease had progressed within less than 24 months.

The CRR results for selected subgroups, based on discussions with clinical experts consulted by CADTH, are presented in Table 16. The results appear to be consistent across all subgroups, with the CRR from |||| ||| || | in patients who received 1 prior line of therapy to ||| ||| || ||| in patients who received 3 prior therapies.

Table 15: Subgroup Analysis of ORR as per Investigator Assessment — 36-Month FAS

Subgroup

ZUMA-5

36-month FAS

N

ORR, n (% [95% CIa])

Number of lines of prior therapy

  1

||

| |||| ||| || |||||

  2

||

|| ||| ||| || ||||

  3

||

|| ||| ||| || ||||

  ≥ 4

||

|| ||| ||| || ||||

Relapsed or refractory

  Relapsed

||

|| ||| ||| || |||||

  Refractory

||

|| ||| ||| || ||||

High tumour bulk per GELF criteria

  Yes

||

|| ||| ||| || ||||

  No

||

|| ||| ||| || ||||

Time to relapse from first anti-CD20 chemotherapy combination therapy

  < 24 months

||

|| ||| ||| || ||||

  ≥ 24 months

||

|| ||| ||| || |||||

CI = confidence interval; FAS = full analysis set; GELF = Groupe d’Étude des Lymphomes Folliculaires; ORR = objective response rate.

Note: ORR is reported as per investigator assessment for the 36-month analysis.

a95% CI is from the Clopper-Pearson method.

Source: ZUMA-5 Clinical Study Report.18

Table 16: Subgroup Analysis of CRR as per Investigator Assessment — 36-Month FAS

Subgroup

ZUMA-5

36-month FAS

N

CRR, n (% [95% CIa])

Number of lines of prior therapy

  1

||

|| |||| ||| || |||||

  2

||

||| || ||| || ||||

  3

||

||| || ||| || ||||

  ≥ 4

||

||| || ||| || ||||

Relapsed or refractory

  Relapsed

||

||| || ||| || ||||

  Refractory

||

||| || ||| || ||||

High tumour bulk per GELF criteria

  Yes

||

||| || ||| || ||||

  No

||

||| || ||| || ||||

Time to relapse from first anti-CD20 chemotherapy combination therapy

  < 24 months

||

||| || ||| || ||||

  ≥ 24 months

||

||| || ||| || ||||

CI = confidence interval; CRR = complete response rate; FAS = full analysis set; GELF = Groupe d’Étude des Lymphomes Folliculaires.

Note: CRR is reported as per investigator assessment for the 36-month analysis.

a95% CI is from the Clopper-Pearson method.

Source: ZUMA-5 Clinical Study Report.18

Duration of Response

A summary of the DOR results from the ZUMA-5 trial is shown in Table 17. At the 36-month FAS time point, ||| of patients no longer demonstrated a response, and the median DOR was 38.6 months (95% CI, 29.0 months to NE). The KM-estimated event-free probability at 18 months was ||||| ||||| || |||||, at 24 months was ||||| ||||| || |||||, and at 36 months was 57.1% (95% CI, 46.6% to 66.3%).

Table 17: DOR in the ZUMA-5 Trial

Characteristic

ZUMA-5

Inferential analysis set

18 months

(N = 81)

FAS

18 months

(N = 117)

FAS

36 months

(N = 119)

Number of patients with events, n (%)

|| ||||

|| ||||

|| ||||

Censored, n (%)a

|| ||||

|| ||||

|| ||||

Time to event (months), median (95% CI)

NE (NE to NE)

|| |||| |||

38.6 (29.0 to NE)

Event-free probability at 18 months, % (95% CI)b

|||| |||||||||||

|||| |||||||||||

|||| |||||||||||

Event-free probability at 24 months, % (95% CI)b

|||| |||||||||||

|||| |||||||||||

|||| |||||||||||

Event-free probability at 36 months, % (95% CI)b

||

||

57.1 (46.6 to 66.3)

Follow-up time (months), median (95% CI)c

|||| |||||||||||

|||| |||||||||||

|||| |||||||||||

CI = confidence interval; FAS = full analysis set; NE = not evaluable; NR = not reported.

Note: DOR is reported according to central assessment at the 18-month analysis and as per investigator assessment for the 36-month analysis.

aReasons for censoring for DOR may include: response ongoing, lost to follow-up, investigator decision, started new anticancer therapy, stem cell transplant, re-treatment.

bSurvival probabilities are according to Kaplan-Meier estimates.

cMedian follow-up time derived using the reverse Kaplan-Meier approach.

Source: ZUMA-5 Clinical Study Report.18

Time to Next Treatment

A summary of the TTNT results from the ZUMA-5 trial is shown in Table 18. At the 36-month FAS time point, ||| of patients had experienced a TTNT event; the median TTNT was NE months (95% CI, 37.8 months to NE). The KM-estimated event-free probability at 18 months was ||||| ||||| || |||||, at 24 months was ||||| ||||| || |||||, and at 36 months was 59.5% (95% CI, 50.2% to 67.6%).

Table 18: TTNT in the ZUMA-5 Trial

Characteristic

ZUMA-5

Inferential analysis set

18 months

(N = 86)

FAS

18 months

(N = 127)

FAS

36 months

(N = 127)

Events, n (%)

|| ||||

|| ||||

|| ||||

Censored, n (%)a

|| ||||

|| ||||

|| ||||

TTNT time (months), median (95% CI)

NE (NE to NE)

|| |||| |||

NE (37.8 to NE)

Event-free probability at 18 months, % (95% CI)b

|||| |||||||||||

|||| |||||||||||

|||| |||||||||||

Event-free probability at 24 months, % (95% CI)b

|||| |||||||||||

|||| |||||||||||

|||| |||||||||||

Event-free probability at 36 months, % (95% CI)b

NR

||

59.5 (50.2 to 67.6)

Follow-up time (months), median (95% CI)c

|||| |||||||||||

|||| |||||||||||

|||| |||||||||||

CI = confidence interval; FAS = full analysis set; NE = not evaluable; NR = not reported; TTNT = time to next treatment.

aReasons for censoring for TTNT may include: alive and without new anticancer therapy, lost to follow-up, withdrawal of consent, investigator decision, and end of study due to other reason.

bSurvival probabilities are according to Kaplan-Meier estimates.

cMedian follow-up time derived using the reverse Kapan-Meier approach.

Source: ZUMA-5 Clinical Study Report.18

Harms

Refer to Table 19 for a summary of harms data in the ZUMA-5 trial.

Adverse Events

At the 36-month time point for analysis, a total of ||| of patients in the safety analysis set experienced a TEAE, with pyrexia (|||||) hypotension (|||), headache (|||), and fatigue (|||) the most frequently reported TEAEs.

Serious Adverse Events

At the 36-month analysis, a total of ||| of patients in the safety analysis set experienced an SAE, with pyrexia (|||||), pneumonia (||||), encephalopathy (||||), and confusional state (||||) the only SAEs reported by at least 5% of patients.

Withdrawals Due to Adverse Events

As axicabtagene ciloleucel is a 1-time infusion, withdrawals due to adverse events are not applicable.

Mortality

At the 36-month time point of analysis, ||| of patients in the safety analysis set had died. The most common reason was due to progressive disease (||), following by AE due to reasons other than progressive disease or subsequent therapy (||) and secondary malignancy (||). Most “other” classifications of death were related to various infections.

Notable Harms

Notable harms identified by CADTH in consultation with the clinical experts consulted on this review included CRS, neurologic events, cytopenias, infection, and hypogammaglobulinemia. At the 36-month analysis, ||| of patients in the safety analysis set had experienced CRS, with || experiencing a grade 3 or higher CRS. Neurologic events were reported in ||| of patients, with ||| reporting a grade 3 or higher neurologic event. Cytopenias were reported in ||| of patients, with ||| of patients reporting a grade 3 or higher cytopenia. Infections were reported in ||| of patients, with ||| reporting a grade 3 or higher infection. Hypogammaglobulinemia was reported in ||| of patients, with || of patients reporting a grade 3 or higher hypogammaglobulinemia.

Table 19: Summary of Harms Results From Studies Included in the Systematic Review

AEs

ZUMA-5

Safety analysis set

18 months

(N = 124)

Safety analysis set

36 months

(N = 124)

Most common AEs, n (%)a

Patients with any TEAE

123 (99)

||| ||||

Pyrexia

103 (83)

||| ||||

Hypotension

59 (48)

|| ||||

Headache

55 (44)

|| ||||

Fatigue

51 (41)

|| ||||

Nausea

45 (36)

|| ||||

Anemia

44 (35)

|| ||||

Neutropenia

|| ||||

|| ||||

Sinus tachycardia

41 (33)

|| ||||

Tremor

36 (29)

|| ||||

Chills

33 (27)

|| ||||

Neutrophil count decreased

|| ||||

|| ||||

Diarrhea

33 (27)

|| ||||

Constipation

35 (28)

|| ||||

Vomiting

29 (23)

|| ||||

Decreased appetite

28 (23)

|| ||||

Hypoxia

27 (22)

|| ||||

Confusional state

28 (23)

|| ||||

Cough

27 (22)

|| ||||

Thrombocytopenia

|| ||||

|| ||||

SAEs, n (%)b

Patients with any SAE

57 (46)

|| ||||

  Pyrexia

16 (13)

|| ||||

  Pneumonia

8 (6)

|| |||

  Encephalopathy

8 (6)

| |||

  Confusional state

7 (6)

| |||

Patients who stopped treatment due to AEs, n (%)

Patients who stopped treatment

||

||

Deaths, n (%)

Patients who died

|| ||||

|| ||||

  Progressive disease

| |||

|| |||

  AE due to reasons other than progressive disease or subsequent therapy

| |||

| |||

  Secondary malignancy

| |||

| |||

  Other, COVID-19

||

| |||

  Other, COVID pneumonia with hypoxic respiratory failure

||

| |||

  Other, infection

| |||

| |||

  Other, lung infection

||

| |||

  Other, sepsis

||

| |||

  Other, Escherichia coli bacteremia/E. coli sepsis with superimposed infection or diarrhea due to Clostridioides difficile infection

||

| |||

  Other, complications of GVHD

||

| |||

  Other, unknown

| |||

| |||

  Other, unknown, found via public record

| |||

| |||

AESIs identified by sponsor, n (%)

CRS

97 (78)

|| ||||

  Grade ≥ 3

8 (6)

| |||

Neurologic event

70 (56)

|| ||||

  Grade ≥ 3

19 (15)

|| ||||

Cytopenias

|| ||||

|| ||||

  Grade ≥ 3

|| ||||

|| ||||

Infection

|| ||||

|| ||||

  Grade ≥ 3

19 (15)

|| ||||

Hypogammaglobulinemia

|| ||||

|| ||||

  Grade ≥ 3

| |||

| |||

AE = adverse event; AESI = adverse event of special interest; CRS = cytokine release syndrome; GVHD = graft vs. host disease; NA = not applicable; SAE = serious adverse event; TEAE = treatment-emergent adverse event.

aPresent in ≥ 20% of patients.

bPresent in > 5% of patients.

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: ZUMA-5 Clinical Study Report.18

Critical Appraisal

Internal Validity

The ZUMA-5 trial, the only eligible study identified by the sponsor, was a phase II, single-arm, open-label clinical trial. The lack of comparative data is a key limitation to the interpretation of the results from a single-arm trial, as it is difficult to distinguish between the effect of the intervention, a placebo effect, or the effect of natural history.54,55 It is acknowledged that there may be practical limitations to conducting a randomized controlled trial in patients with r/r FL (beyond first-line treatment), such as decreasing population size with subsequent lines of therapy and lack of a standard treatment in these later lines of treatment. However, the sponsor’s submission included information on the phase III ZUMA-22 trial,56 which is studying axicabtagene ciloleucel compared to standard of care, and which is currently recruiting patients with results expected in 2027;56 this suggests that it is possible to conduct a phase III comparative trial in this population. The hypothesized historical control of 40% ORR and 15% CRR was used to determine a clinically meaningful benefit. The prespecified thresholds were established against the response rate derived from studies of available PI3K inhibitors for the treatment of r/r FL.57 The clinical experts consulted by CADTH supported using a CRR of 15% and an ORR of 40% as clinically relevant thresholds.

Due to the open-label design of the trial, the response outcomes measures (i.e., ORR, DOR, PFS) and subjective harms are at risk of measurement or reporting bias, though the direction of this bias is unclear. The primary end point of ORR was assessed by central assessment at the 18-month primary analysis, which reduces the likelihood that the open-label nature of the trial impacted ORR. Although response rates at the 36-month time point were only assessed by the investigator, which can impact reporting of AEs, response rates as measured by central and investigator assessments were similar at the 18-month analysis, which suggests accurate reporting of the ORR by investigators.

Follow-up time was deemed sufficient for assessing tumour response and safety outcomes associated with axicabtagene ciloleucel. The clinical experts consulted by CADTH noted that r/r FL is a disease that can have very long periods of PFS and survival, suggesting that the follow-up duration was not long enough to fully capture the effects on OS and PFS. In addition to the duration of the study and the noncomparative design, subsequent treatments make it difficult to interpret the OS results. After the infusion of axicabtagene ciloleucel, ||| of the patients received at least 1 subsequent antineoplastic medication, and ||| of them received SCT. The estimated clinical end point curve (OS) should be interpreted in cases where subsequent treatments are given as the magnitude of patient benefit due to treatment is difficult to quantify.

As treatment with axicabtagene ciloleucel is a 1-time infusion, adherence to treatment is not a concern for internal validity. Of the 127 patients included in the 36-month FAS, ||| patients (|||) had discontinued the study before receiving axicabtagene ciloleucel. Clinical experts consulted by CADTH considered this to be a realistic proportion of patients who are unable to complete the treatment process, and representative of clinical practice.

External Validity

According to the clinical experts consulted by CADTH, the ZUMA-5 study population overall represents the patients in the population with r/r FL in Canada who would be receiving axicabtagene ciloleucel. However, the clinical experts noted that patients seen in clinical practice would include those with poorer performance status (the ZUMA-5 trial only included patients with an ECOG PS score of 0 or 1, whereas clinical experts suggest that patients with an ECOG PS score of 2 may be treated in the clinical setting), as well as patients with more comorbidities. The clinical experts differed in their opinions regarding patients who received prior CD19-targeted therapy; some suggested that any prior CD19-targeted therapy would preclude the use of axicabtagene ciloleucel, whereas others suggested that only patients who were refractory to CD19-targeted therapy (did not respond or relapsed within 6 months) would not be suitable candidates for treatment with axicabtagene ciloleucel.

After screening, the procedures and co-interventions (including manufacturing process, depleting chemotherapy, bridging therapy, and post–axicabtagene ciloleucel interventions) were consistent with those adopted in the Canadian setting, although some minor discrepancies exist. The ZUMA-5 trial results can nevertheless be generalized to the population of patients in Canada.

Subgroup analyses were conducted based on prespecified patient characteristics. Due to insufficient follow-up time to observe a large number of survival end points, the OS and PFS subgroup analyses were considered unstable and therefore uninformative. Based on the subgroup analyses conducted for ORR and CRR, efficacy appears to be present for each patient population based on prior therapies, POD24, tumour bulk, or r/r status.

According to the clinical experts consulted by CADTH, the efficacy outcomes used in this study are clinically relevant and important for the clinical trials in r/r FL, with the notable exception of HRQoL outcomes, which are important to patients but were excluded from the ZUMA-5 trial. As such, it is not possible to determine how the introduction of axicabtagene ciloleucel will impact the HRQoL of patients in Canada.

Lack of long-term data on patients’ survival and response rates is another limitation, given that FL is an indolent and slowly progressing disease. Clinical benefits of the treatment need to be evaluated over a longer follow-up time to increase confidence in the durability of response expected from the use of axicabtagene ciloleucel in Canada.

Studies Addressing Gaps in the Systematic Review Evidence

Contents within this section have been informed by materials submitted by the sponsor. The following has been summarized and validated by the CADTH review team.

Objectives for the Summary of Other Relevant Evidence

The ZUMA-5 trial is a single-arm trial and therefore does not provide evidence of efficacy against standard of care for patients with r/r FL who have received 2 or more lines of therapy. Treatment choice for r/r FL is dependent on a variety of factors, including prior therapies, duration of remission, patient-related factors such as age, and clinician/patient preferences. As such, the comparator patients receive can vary considerably. The sponsor submitted evidence of relative efficacy of axicabtagene ciloleucel against standard of care therapies in patients with r/r FL who have received 2 or more lines of therapy.

Description of Other Relevant Evidence

The relative efficacy of axicabtagene ciloleucel versus standard of care was estimated in the ZUMA-5 population using propensity scores with SMR weights.19 SMR weighting estimates the treatment effect in a population with an equal distribution of risk factors to that of the treatment study participants only.58 SCHOLAR-5, the standard of care cohort, is a retrospective, observational, multicentre, database study of patients with r/r FL (grades 1, 2, or 3a) who have received 2 or more systemic therapies. The SCHOLAR-5 cohort was derived from 3 international cohorts: IQVIA, Vanderbilt, and DELTA. The IQVIA and Vanderbilt cohorts were created from electronic medical records, while the DELTA cohort represented patients from the DELTA clinical trial (NCT01282424) who proceeded to receive therapy after idelalisib treatment. The DELTA cohort was added to increase statistical power for OS, TTNT, and response outcomes. However, the DELTA cohort was not used to inform PFS as tumour assessment dates were not provided for the line of therapy subsequent to idelalisib (i.e., the index line of therapy). The index line of therapy was chosen at random from all lines of therapy received by a patient after they met all the eligibility criteria for the study. The index date for the primary analysis was defined as the initiation date of the patient’s index line. A summary of the SCHOLAR-5 study is provided in Table 20.

Table 20: Study Selection Criteria and Methods for Other Relevant Evidence Submitted by the Sponsor

Characteristics

Indirect comparison

Population

Patients with r/r grade 1, 2, or 3a FL after 2 or more systemic therapies

Interventions

Any therapy used for r/r grade 1, 2, or 3a FL in the third- and later-line treatment setting

Comparator

Standard of care therapies:

  • Anti-CD20 agent + alkylating chemotherapy

  • Alkylating chemotherapy

  • Allo-SCT or auto-SCT

  • Experimental therapy

  • Bruton tyrosine kinase inhibitor

  • PI3K inhibitor

  • Immunomodulatory agent

  • Fludarabine

  • Radioimmunotherapy

Outcome

  • OS: time from index date to death.

  • PFS: time from index date until earliest date of progression or death from any cause.

  • TTNT: time from index date to initiation of next therapy or death.

  • DOR: time from first objective response within the line of therapy until disease progression or death due to any cause, whichever comes first. DOR is only defined for patients with a PR or CR.

  • ORR: proportion of patients achieving either a CR or PR as indicated by direct documentation in the patient’s medical record since the index date.

Study designs

Retrospective, observational study

Data used

Data on file

Inclusion criteria

  • Patients aged ≥ 18 years

  • Patients with histologically confirmed diagnosis of iNHL, with histological subtype limited to FL grade 1, 2, or 3a, based on criteria established by the WHO 2016 classification

  • Patients with r/r disease starting third or later line of therapy on or after July 23, 2014a

Exclusion criteria

  • Transformed FLb

  • FL histological grade 3b

  • Prior CAR T-cell therapy or other genetically modified T-cell therapy

  • Eligible within 12 months before the last updated version of the database

  • ECOG PS score > 1

  • CNS involvement

allo-SCT = allogeneic stem cell transplant; auto-SCT = autologous stem cell transplant; CNS = central nervous system; CR = complete response; DOR = duration of response; ECOG PS = Eastern Cooperative Oncology Group Performance Status; FL = follicular lymphoma; iNHL = indolent non-Hodgkin lymphoma; ORR = objective response rate; OS = overall survival; PFS = progression-free survival; PI3K = phosphoinositide 3-kinase; PR = partial response; r/r = relapsed or refractory; TTNT = time to next treatment.

aEligibility criteria were not restricted by date for patients from the DELTA clinical trial.

bPatients who pass all other inclusion/exclusion criteria but transform at a later date were eligible until transformation.

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: Sponsor-provided analysis report.19

Analysis Methods

Patient-level data for the ZUMA-5 and SCHOLAR-5 studies were used to inform a comparative analysis. Propensity scores were calculated for each patient in the pooled analysis set to account for differences in baseline characteristics across populations. These calculated scores were then used to apply SMR weighting, with differences required to be less than 0.1. All ZUMA-5 patients were included in the analysis. Selection of variables for the propensity score model was determined in a hierarchal manner and based on the advice of investigators or clinical experts, with the goal of minimizing the imbalance in prognostically important covariates. For high and medium priority rank variables with less than 40% missing data, multiple imputation was performed.

The variables included in the propensity score model were:

Weighted logistic regression was used to estimate ORs of the ORR and CRR across cohorts. A weighted KM estimator of the risk probabilities and 95% CIs for OS, PFS, TTNT, and DOR were estimated at 3-month intervals.19 Time-to-event outcomes were summarized via hazard ratios, estimated using a Cox proportional hazard model. Statistical sensitivity analyses of ORR and OS were conducted to support the robustness of the findings. Additional sensitivity analyses performed for all outcomes included: exclusion of DELTA trial patients from the SCHOLAR-5 cohort, using the date of axicabtagene ciloleucel infusion as the start point for time-to-event variables, and using the safety and inferential analysis sets. Prespecified subgroup analyses included investigations on patient disease status and prior therapies. A summary of the methods is shown in Table 21.

Table 21: Analysis Methods

Methods

Description

Balancing methodology

Propensity score weighting: A logistic regression model was used to estimate propensity scores. Weights, assigned in the SMR method, were calculated for individuals in the SCHOLAR-5 cohort as: propensity score / (1–propensity score).

PSM: Performed as a sensitivity analysis, patients in the ZUMA-5 trial were matched with a patient in the SCHOLAR-5 study exhibiting the nearest propensity score.

Propensity score specification

Propensity scores were specified by patient characteristics in a hierarchal order of importance, based on investigator and clinical advice.

Covariates included

  • POD24 after initiation of first-line anti-CD20 chemotherapy combination therapy

  • Number of lines of prior therapy

  • Relapsed vs. refractory at index

  • Prior SCT

  • Time from last treatment

  • Best response to last line of therapy

  • Tumour bulk (diameter of largest lesion)

  • Age

  • Prior anti-CD20 + alkylating agent

Multiple imputation

Multiple imputation was applied for variables with missing data (< 40%) that were specified as part of the propensity score model. Little’s test of missing completely at random was performed, which provided a significance value of P < 0.001.

Variables that required imputation were:

  • tumour bulk

  • time since last treatment

  • CR or PR to prior line of therapy.

Outcomes

  • ORR

  • OS (date of leukapheresis as start point)

  • PFS (date of leukapheresis as start point)

  • TTNT (date of leukapheresis as start point)

  • DOR (date of first objective response as start point)

  • Best overall response (CR, PR, stable disease)

Follow-up time points

ZUMA-5: 36-month FAS; the SCHOLAR-5 study is retrospective study where at least 12 months of follow-up was required

Model estimation

Two-sided CI was utilized, and all tests were performed on the 5% alpha level.

For time-to-event variables, HRs of the outcome between groups were estimated using a Cox proportional hazard model.

Sensitivity analyses

For ORR, the robustness of findings was tested using 3 types of bootstrap CI (computed using the percentile method, normal distribution method, and bias-corrected method) and the robust Wald assessment. Additional sensitivity analyses were performed using inverse probability of treatment weights for doubly robust analysis and nullification analysis for assessment of unmeasured confounders and their association with the outcome.

For OS, a statistical sensitivity analysis was prespecified via the parametric g-formula to assess differences between the ZUMA-5 and SCHOLAR-5 groups.

For all outcomes, additional sensitivity analyses included:

  • PSM analysis (as opposed to PS weighting)

  • Propensity score unweighted analysis (as opposed to propensity score weighting)

  • Exclusion of DELTA trial patients from the SCHOLAR-5 cohort. This sensitivity analysis was performed to account for the involvement of DELTA trial patients in a clinical trial, which was not the case for SCHOLAR-5 patients

  • Analysis using the ZUMA-5 safety and inferential analysis sets

  • Date of axicabtagene ciloleucel infusion as the start point for time-to-event variables (PFS, OS, TTNT), as opposed to date of leukapheresis.

Subgroup analysis

The comparative effectiveness of axicabtagene ciloleucel vs. standard of care was assessed in prespecified subgroups of interest as follows:

  • Patients with POD24 after initiation of first-line anti-CD20 chemotherapy combination therapy

  • Patients who were refractory at index date

  • Patients who had failed 3 or more lines of therapy.

CI = confidence interval; CR = complete response; DOR = duration of response; FAS = full analysis set; HR = hazard ratio; ORR = overall response rate; OS = overall survival; PFS = progression-free survival; POD24 = progression of disease within 24 months; PR = partial response; PS = propensity score; PSM = propensity score matching; SCT = stem cell transplant; SMR = standardized mortality ratio; TTNT = time to next treatment; vs. = versus.

Note: Low priority variables were not included in the propensity score weighting.

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: Sponsor-provided analysis report.19

Results

Summary of Included Studies

The comparative analysis contrasted 2 populations, those in the ZUMA-5 and SCHOLAR-5 studies, to derive estimates of relative efficacy for axicabtagene ciloleucel and standard of care. The ZUMA-5 trial is a single-arm, phase II study evaluating the efficacy and safety of axicabtagene ciloleucel in patients with r/r grade 1, 2, or 3a FL (n = 127). The SCHOLAR-5 cohort was constructed retrospectively from 3 international observational cohorts: IQVIA, Vanderbilt, and DELTA. The DELTA cohort included patients from the DELTA clinical trial (NCT01282424) who proceeded to receive therapy after idelalisib treatment. The DELTA cohort in the SCHOLAR-5 trial did not report progression assessment dates and had to be removed from the PFS and DOR analysis. Table 22 summarizes the assessment of homogeneity between the ZUMA-5 trial and the SCHOLAR-5 cohort.

Table 22: Assessment of Homogeneity

Characteristics

Description and handling of potential effect modifiers

Disease severity

Disease severity was similar across the SCHOLAR-5 and ZUMA-5 studies as both studies required patients to have a confirmed diagnosis of FL (grade 1, 2, or 3a) with r/r disease and starting their third (or later) line of treatment. Prior to weighting, the proportion of patients in the SCHOLAR-5 study with POD24 was smaller (35.7%) than the proportion of patients in the ZUMA-5 trial with POD24 (55.1%).

Treatment history

Patients in the SCHOLAR-5 study had fewer lines of prior therapy (mean of |||| | ||||) compared to patients in the ZUMA-5 trial (|||| | ||||).

Eligibility criteria

Eligibility criteria for the SCHOLAR-5 study was aligned with ZUMA-5 criteria. Patients in both studies were ≥ 18 years and had histologically confirmed iNHL limited to FL (grade 1, 2, or 3a) based on WHO 2016 classification.48 Eligible patients also had r/r disease and were starting their third (or later) line of therapy. Patients were excluded if they had transformed FL or FL histological grade 3b. Additional exclusion criteria were prior CAR T-cell therapy or other genetically modified T-cell therapy, ECOG PS score > 1, and involvement of the CNS. A subset of eligible patients from the DELTA clinical trial were included to increase statistical power for OS, TTNT, and response rates.

Dosing of comparators

In the ZUMA-5 study, patients received 2 × 106 anti-CD19 CAR T cells/kg body weight. The SCHOLAR-5 study involved patients who received any available treatment for r/r FL, including approved and experimental therapies, and autologous and allogeneic transplant. The index LOT for SCHOLAR-5 participants was randomly selected from eligible LOTs. For the subset of patients derived from the DELTA trial, the index LOT was the treatment received after idelalisib. Ineligible index treatments for the SCHOLAR-5 study included single-agent anti-CD20 therapy, surgery, and radiotherapy alone. CAR T-cell therapy and other cellular therapies were also ineligible index treatments.

Definitions of end points

The SCHOLAR-5 study defined ORR as the proportion of patients achieving either CR or PR, as indicated by direct documentation in the patient’s medical record since the index date. OS, PFS, and TTNT were defined as the time from index date to death (in the case of OS), progression or death (in the case of PFS), or initiation of next therapy or death (in the case of TTNT). In the ZUMA-5 trial, the start point date for these time-to-event outcomes was date of leukapheresis. DOR was defined as the time from first objective response within the line of therapy until disease progression or death.

Timing of end point evaluation

Efficacy end points as described above were measured at the 36-month follow-up analysis in the ZUMA-5 trial. The SCHOLAR-5 study is a retrospective study where at least 12 months of follow-up was required. Time-to-event end points (OS, PFS, TTNT, DOR) were also compared at 3-month intervals.

Clinical trial setting

The SCHOLAR-5 cohort was generated from database records provided by IQVIA and Vanderbilt University Medical Center. The SCHOLAR-5 study also included eligible patients from the DELTA clinical trial. Data from the ZUMA-5 trial came from patients who were assessed at 17 investigative sites across the US and France.

Study design

The ZUMA-5 trial is a single-arm, open-label, phase II study. The SCHOLAR-5 study is a retrospective database study, acquiring data from multiple centres and from the DELTA clinical trial.

CAR = chimeric antigen receptor; CNS = central nervous system; CR = complete response; DOR = duration of response; ECOG PS = Eastern Cooperative Oncology Group Performance Status; iNHL = indolent non-Hodgkin lymphoma; FL = follicular lymphoma; LOT = line of therapy; ORR = ; OS = overall survival; PFS = progression-free survival; POD24 = progression of disease within 24 months; PR = partial response; r/r = relapsed or refractory; TTNT = time to next treatment.

Details included in this table are from the sponsor’s Summary of Clinical Evidence.59

Source: Sponsor-provided analysis report.19

Table 23 summarizes the index treatment patterns in the SCHOLAR-5 cohort before propensity score weighting. The most common index treatments, in descending order, were anti-CD20 agent plus bendamustine (16.8%), experimental therapy (16.1%), immunomodulatory agent (12.6%), PI3K inhibitor (9.8%), auto-SCT (8.4%), anti-CD20 agent plus other chemotherapy (8.4%), and other chemotherapy (8.4%).

A full description of the ZUMA-5 trial can be found in the Systematic Review section of this report.

Table 23: Index Treatment Patterns for the SCHOLAR-5 (n = 143) Sample of Patients With 2 or More Lines of Prior Therapy

Treatment

Frequency, n (%)

Alkylating chemotherapy

1 (0.7)

Allo-SCT

4 (2.8)

Auto-SCT

12 (8.4)

Bruton tyrosine kinase inhibitor

1 (0.7)

Anti-CD20 agent + alkylating chemotherapy

5 (3.5)

Anti-CD20 agent + bendamustine

24 (16.8)

Anti-CD20 agent + CHOP-like chemotherapy

7 (4.9)

Anti-CD20 agent + fludarabine-based chemotherapy

3 (2.1)

Anti-CD20 agent + immunomodulatory agent

1 (0.7)

Anti-CD20 agent + platinum-based chemotherapy

1 (0.7)

Anti-CD20 agent + other chemotherapy

12 (8.4)

Other chemotherapy

12 (8.4)

Experimental therapy

23 (16.1)

Fludarabine

1 (0.7)

Immunomodulatory agent

18 (12.6)

PI3K inhibitor

14 (9.8)

Radioimmunotherapy

2 (1.4)

SCT (other)

2 (1.4)

allo-SCT = allogeneic stem cell transplant; auto-SCT = autologous stem cell transplant; CHOP = cyclophosphamide + doxorubicin-vincristine-prednisone; PI3K = phosphoinositide 3-kinase; SCT = stem cell transplant.

Source: Sponsor-provided analysis report.19

Results — Propensity Score Weighting

The SCHOLAR-5 cohort was reweighted using the propensity scores to further align with the ZUMA-5 patient population. After propensity score weighting, the SCHOLAR-5 effective sample size was reduced from 143 patients to 128 patients. The largest difference between cohorts was in time from last treatment; after weighting the average was reduced from the original ||||| months in the full SCHOLAR-5 population to |||| months, and the proportion of patients with POD24 increased from 35.7% in the unadjusted population to 57.1% in the propensity score–weighted population. After propensity score weighting, variables were mostly similar to the ZUMA-5 population. The removal of the DELTA cohort from the SCHOLAR-5 cohort also resulted in relatively balanced variables; however, the proportion of patients with POD24 and the proportion who were refractory to their most recent treatment were both lower.

A summary of the patient characteristics not included in the propensity score model for both the ZUMA-5 and SCHOLAR-5 trials before and after weighting is shown in Table 25. The largest differences between the post-weighting SCHOLAR-5 and ZUMA-5 trials are in the proportion of patients with an ECOG PS score of 0, that is, 32.6% versus 62.2%, respectively. Other large differences between the SCHOLAR-5 and ZUMA-5 trials are in the proportion of patients who are double refractory (||||| versus |||||, respectively) and have bone marrow involvement (||||| versus |||||, respectively).

Table 24: Propensity Score Variables in the ZUMA-5 and SCHOLAR-5 Trials

Propensity score variables

SCHOLAR-5

ZUMA-5

(N = 127)

P value

SMD

Before weighting

(N = 143)

After weighting

(N = 128)

POD24, n (%)

51 (35.7)

73 (57.1)

70 (55.1)

0.789

0.039

Number of lines of prior therapy, mean (SD)

|||| ||||||

|||| ||||||

|||| ||||||

|||||

|||||

Relapsed or refractory to prior line of therapy, n (%)

  Relapsed

57 (39.5)

36 (25.5)

40 (31.5)

0.560

0.083

  Refractory

86 (60.5)

93 (72.3)

87 (68.5)

Prior SCT, n (%)

31 (21.7)

33 (25.5)

30 (23.6)

0.783

0.043

Tumour bulk (cm), mean (SD)

|||| ||||||

|||| ||||||

|||| ||||||

|||||

|||||

Time since last treatment (months), mean (SD)

||||| |||||||

|||| |||||||

|||| |||||||

|||||

|||||

Response to prior line of therapy, n (%)

  CR

|| ||||||

|| ||||||

|| ||||||

|||||

|||||

  PR

|| ||||||

|| ||||||

|| ||||||

  Stable disease

|| ||||||

|| ||||||

|| ||||||

  Progressive disease

|| ||||||

|| ||||||

|| ||||||

Age, mean (SD)

||||| |||||||

||||| |||||||

||||| ||||||

|||||

|||||

Prior anti-CD20 + alkylator combination treatment, n (%)

||| ||||||

||| |||||

||| |||||

|||||

||||||

Propensity score variables, after weighting (removal of DELTA cohort)

N (%)

NA

|| ||||||

||| |||||

|||

|||

POD24, n (%)

NA

|| ||||||

|| ||||||

|||||

|||||

Number of lines of prior therapy, mean (SD)

NA

|||| ||||||

|||| ||||||

|||||

|||||

Relapsed or refractory to prior line of therapy, n (%)

||

||

  Relapsed

NA

|| ||||||

|| ||||||

|||||

|||||

  Refractory

NA

|| ||||||

|| ||||||

Prior SCT, n (%)

NA

|| ||||||

|| ||||||

|||||

|||||

Tumour bulk (cm), mean (SD)

NA

|||| ||||||

|||| ||||||

|||||

|||||

Time since last treatment (months), mean (SD)

NA

||||| |||||||

|||| |||||||

|||||

|||||

Response to prior line of therapy, n (%)

||

  CR

NA

|| ||||||

|| ||||||

|||||

|||||

  PR

NA

|| ||||||

|| ||||||

  Stable disease

NA

|| ||||||

|| ||||||

  Progressive disease

NA

|| ||||||

|| ||||||

Age, mean (SD)

NA

||||| |||||||

||||| ||||||

|||||

|||||

Prior anti-CD20 + alkylator combination treatment, n (%)

NA

|| |||||

||| |||||

|||||

||||||

CR = complete response; POD24 = progression of disease within 24 months; PR = partial response; SCT = stem cell transplant; SD = standard deviation; SMD = standardized mean difference.

Source: Sponsor-provided analysis report.19

Table 25: Patient Characteristics not Included in the Propensity Score Model in the ZUMA-5 and SCHOLAR-5 Trials

Propensity score variables

SCHOLAR-5

ZUMA-5

(N = 127)

P value

SMD

Before weighting

(N = 143)

After weighting

(N = 128)

FL subtype, n (%)

  Grade 1

|| ||||||

|| ||||||

|| ||||||

|||||

|||||

  Grade 2

|| ||||||

|| ||||||

|| ||||||

  Grade 3a

|| ||||||

|| ||||||

|| ||||||

  Missing

|| |||||

| |||||

| |||

ECOG PS, n (%)

  0

39 (33.1)

35 (32.6)

79 (62.2)

< 0.001

0.621

  1

79 (66.9)

72 (67.4)

48 (37.8)

  Missing

|| ||||||

|| ||||||

| |||

Double refractory, n (%)

|| ||||||

|| ||||||

|| ||||||

|||||

|||||

Prior radiation, n (%)

|| ||||||

|| ||||||

||| |||||

||||||

|||||

Prior alkylating monotherapy, n (%)

||| ||||||

||| |||||

|| ||||||

||||||

|||||

Prior anti-CD20 monotherapy, n (%)

||| |||||

||| |||||

|| ||||||

||||||

|||||

Prior lenalidomide, n (%)

| |||||

| |||||

|| ||||||

||||||

|||||

Prior PI3K inhibitor, n (%)

|| ||||||

|| ||||||

|| ||||||

|||||

|||||

Bone marrow involvement, n (%)

|| ||||||

|| ||||||

|| ||||||

|||||

|||||

FLIPI, n (%)

  0

| |||||

| |||||

| |||||

|||||

|||||

  1

| |||||

| ||||||

|| ||||||

  2

|| ||||||

| ||||||

|| ||||||

  3

|| ||||||

|| ||||||

|| ||||||

  4

|| ||||||

| ||||||

|| ||||||

  5

| |||||

| ||||||

| |||||

  Missing

|| ||||||

|| ||||||

| |||

Hemoglobin (g/dL), mean (SD)

||||| |||||

||||| ||||||

12.60 (1.92)

0.027

0.363

ECOG PS = Eastern Cooperative Oncology Group Performance Status; FL = follicular lymphoma; FLIPI = Follicular Lymphoma Internal Prognostic Index; PI3K = phosphoinositide 3-kinase; SD = standard deviation; SMD = standardized mean difference.

Source: Sponsor-provided analysis report.19

A summary of the estimated relative efficacy is provided in Table 26. The ORR in the ZUMA-5 population was 93.7% compared to 54.0% in the propensity score–weighted SCHOLAR-5 population (OR = 12.66; 95% CI, 5.24 to 30.57; P < 0.001). The CRR in the ZUMA-5 population was 78.7% compared to 34.9% in the propensity score–weighted SCHOLAR-5 population (OR = 6.90; 95% CI, 3.62 to 13.18; P < 0.001).

The median PFS in the ZUMA-5 population was 40.21 months (95% CI, 28.94 months to NE) compared to 12.97 months (95% CI, 7.75 months to 15.47 months) in the propensity score–weighted SCHOLAR-5 population, with HR of 0.27 (95% CI, 0.18 to 0.41; P < 0.0001. The KM curve for the PFS analysis is shown in Figure 4. The median OS in the ZUMA-5 population was NE (95% CI, NE to NE) compared to NE (95% CI, 38.40 months to NE) in the propensity score–weighted SCHOLAR-5 population (HR = 0.56; 95% CI, 0.33 to 0.95; P = 0.0303). The KM curve for the OS analysis is shown in Figure 5. The median TTNT in the ZUMA-5 population was NE (95% CI, 37.85 months to NE) compared to 26.61 months (95% CI, 12.65 months to NE) in the propensity score–weighted SCHOLAR-5 population (HR = 0.25; 95% CI, 0.15 to 0.41; P < 0.0001).

The median DOR in the ZUMA-5 population was 38.64 months (95% CI, 29.04 months to NE) compared to ||||| |||||| ||||| || |||||| in the propensity score–weighted SCHOLAR-5 population, with HR of |||| ||||| || |||||; P ||||||.

Table 26: Summary of Estimated Relative Efficacy in r/r FL

Measure

Propensity score–weighted analysis

SCHOLAR-5

(N = 128)

ZUMA-5 36-month analysis

(N = 127)

ORR

n (%)

69 (54.0)

119 (93.7)

OR (95% CI)

12.66 (5.24 to 30.57)

P value

< 0.001

CRR

n (%)

45 (34.9)

100 (78.7)

OR (95% CI)

6.90 (3.62 to 13.18)

P value

< 0.001

OS

n

128

127

OS time (months), median (95% CI)

NE (38.40 to NE)

NE (NE to NE)

Cox model HR (95% CI)

0.56 (0.33 to 0.95)

P value

0.0303

G-estimation HR (95% CI)

|||| |||||| |||||

P value

||||||

PFS

n

89

127

PFS time (months), median (95% CI)

12.97 (7.75 to 15.47)

40.21 (28.94 to NE)

Cox model HR (95% CI)

0.27 (0.18 to 0.41)

P value

< 0.0001

TTNT

n

128

127

TTNT time (months), median (95% CI)

26.61 (12.65 to NE)

NE (37.85 to NE)

Cox model HR (95% CI)

0.60 (0.39 to 0.93)

P value

0.0223

DOR

n

||

|||

DOR time (months), median (95% CI)

||||| |||||| ||||||

38.64 (29.04 to NE)

Cox model HR (95% CI)

|||| |||||| |||||

P value

|||||||

CI = confidence interval; CRR = complete response rate; DOR = duration of response; HR = hazard ratio; NE = not evaluable; OR = odds ratio; ORR = objective response rate; OS = overall survival; PFS = progression-free survival; TTNT = time to next treatment.

Note: OR > 1 and HR < 1 demonstrate a relative benefit of axicabtagene ciloleucel vs. the comparator basket.

Source: Sponsor-provided analysis report.19

Figure 4: PFS in the ZUMA-5 Trial (36-Month FAS) Compared to the SCHOLAR-5 Study (Propensity Score Weighted)

The figure shows the Kaplan-Meier estimate curves for PFS, with proportion shown on the y-axis and time, in months, on the x-axis. The ZUMA-5 curve is above the SCHOLAR-5 curve from time point 0. At time point 0, both curves are at 100% on the y-axis. The SCHOLAR-5 curve is much steeper, dropping to 0% at 36 months. The ZUMA-5 curve is less steep, dropping to about 50% at about 48 months.

FAS = full analysis set; PFS = progression-free survival.

Source: Sponsor-provided analysis report.19

Figure 5: OS in the ZUMA-5 Trial (36-Month FAS) Compared to the SCHOLAR-5 Trial (Propensity Score Weighted)

The figure shows the Kaplan-Meier estimate curves for overall, with proportion of the patient population shown on the y-axis and time, in months, on the x-axis. At time point 0, both the ZUMA-5 and the SCHOLAR-5 curves are at 100% on the y-axis. The SCHOLAR-5 curve descends to about 50% at time point 60 months and then plateaus for the remaining time to 69 months. The ZUMA-5 curve is slightly less steep, staying above the SCHOLAR-5 curve from time point 0, and dropping to about 75% at about 39 months, and then plateauing for the remaining time to 57 months.

FAS = full analysis set; OS = overall survival.

Source: Sponsor-provided analysis report.19

Critical Appraisal of Other Relevant Evidence

Internal Validity

The submitted comparative analysis provides an estimate of relative efficacy of axicabtagene ciloleucel, using evidence from the ZUMA-5 clinical trial, against a basket of therapies representing standard of care, using evidence from the SCHOLAR-5 cohort, a combination of retrospective real-world evidence and a subset of the DELTA clinical trial.

Due to differences between the ZUMA-5 and SCHOLAR-5 cohorts in treatment allocation, it is possible that the treatment effect estimate is confounded by imbalances in prognostic covariates across populations. The sponsor identified and adjusted for several important variables, resulting in a suitable balance of these characteristics across both populations. However, characteristics deemed critical by the clinical experts — FLIPI score, ECOG PS score, and double refractory status — were omitted; FL grade was also omitted. Of the variables collected in both cohorts, there were observed imbalances across treatment groups even after reweighting the populations. The variables were acknowledged by the sponsor, but were omitted due to concerns regarding missing data. The clinical experts consulted by CADTH suggested that differences in ECOG PS scores and the proportion of patients who are double refractory could affect how patients would be expected to respond to treatment. The direction of this impact is uncertain, with some differences potentially favouring axicabtagene ciloleucel over the SCHOLAR-5 comparator. There is additional uncertainty in the results because of the low effective sample sizes in both the ZUMA-5 trial and the SCHOLAR-5 cohort. The removal of the DELTA cohort resulted in a statistically significant change in the mean number of lines of prior therapy, that is, |||| |||||| in the SCHOLAR-5 study compared to |||| |||||| in the ZUMA-5 trial. Differences between populations in the number of lines of prior therapy have a particularly large impact when determining how patients would be expected to respond to treatment. The proportion of patients with POD24 and the proportion of patients who were refractory to their most recent treatment were also both reduced with the exclusion of the DELTA cohort, indicating that the removal of this cohort results in a population with a lower risk prognosis. The ZUMA-5 trial and the SCHOLAR-5 study had different follow-up time requirements: the ZUMA-5 cohort used a 36-month follow-up, while the SCHOLAR-5 cohort required at least 12 months of follow-up. It is possible that individuals’ characteristics or issues with treatment adherence determined patients’ decisions to exit the cohort early and introduced informative censoring, which bias the results.

External Validity

The clinical experts confirmed that the distribution of therapies in the SCHOLAR-5 cohort were representative of the standard of care for patients in Canada; however, it is unclear how this distribution was affected by the propensity score weighting or the exclusion of the DELTA cohort, as the comparator therapies used were not reported in the post-weighting population. The inclusion and exclusion criteria for the SCHOLAR-5 cohort was similar to the ZUMA-5 trial, although it is uncertain if the results can be generalized beyond this selected group of patients.

Future Planned Studies

Contents within this section have been informed by materials submitted by the sponsor. The following have been summarized and validated by the CADTH review team.

Table 27: Summary of Gaps in the Systematic Review Evidence

Evidence gap

Studies that address gaps

ZUMA-22

Summary of key results

  • Patients with POD24 were not included in the ZUMA-5 trial.

  • HRQoL was not assessed in the pivotal study.

A prospective, interventional, open-label, randomized, phase III study assessing the efficacy and safety of axicabtagene ciloleucel vs. standard of care in patients with r/r FL after 2 or more lines of systemic therapy, or r/r FL after first-line chemoimmunotherapy in patients with POD24.

NA

FL = follicular lymphoma; HRQoL = health-related quality of life; NA = not applicable; POD24 = progression of disease within 24 months; r/r = relapsed or refractory.

Source: Sponsor’s Summary of Clinical Evidence.59

Description of Studies

The ZUMA-22 trial56 is a prospective, interventional, open-label, randomized, phase III study assessing the efficacy and safety of axicabtagene ciloleucel versus standard of care in patients with r/r FL after 2 or more lines of systemic therapy or r/r FL after first-line chemoimmunotherapy in patients with POD24. The study began in September 2022 and is actively recruiting participants. Final study results of the ZUMA-22 trial are expected by Health Canada in |||| as a requirement for the issued Notice of Compliance for adult patients with r/r grade 1, 2, or 3a FL after 2 or more lines of systemic therapy.

Discussion

Summary of Available Evidence

One clinical trial was included in the systematic review conducted by the sponsor. The ZUMA-5 study is a phase II, open-label, single-arm study that evaluated the efficacy and safety of axicabtagene ciloleucel in patients with r/r grade 1, 2, or 3a FL after 2 or more lines of systemic therapy. The primary end point was ORR per central assessment in the inferential analysis set at the 18-month time point. Secondary end points included CRR, OS, PFS, DOR, and TTNT. A total of 127 patients with r/r FL were enrolled, with 86 included in the 18-month primary analysis. Data for the full analysis up to the 36-month time point were available at the time of this review (the data cut-off was March 31, 2022). The median age in the FAS was 60 years (range, 34 years to 79 years). More males (59%) were enrolled than females (41%). Most patients had an ECOG PS score of 0 (62%), and the most common number of prior therapies was 2 (|||||). The proportion of patients with POD24 was 55%.

The sponsor provided an additional study in which patient-level data from the retrospective SCHOLAR-5 study were reweighted using propensity scores to be comparable with the ZUMA-5 populations. Given that the ZUMA-5 trial is a single-arm noncomparative study, adjusting the SCHOLAR-5 patient population to be more similar to the ZUMA-5 population allows for an estimate of comparative efficacy against standard of care within the ZUMA-5 population. The outcomes analyzed were ORR, CRR, DOR, OS, PFS, and TTNT. The treatments that made up the basket of therapies in the SCHOLAR-5 study were any available treatment for r/r FL, including approved and experimental therapies, and autologous and allogeneic transplant.

Interpretation of Results

Efficacy

In the ZUMA-5 trial, based on the primary end point ORR, 94% (95% CI, 88% to 97%) of patients achieved a partial response to axicabtagene ciloleucel treatment 36 months following infusion, and based on the secondary end point CRR, 79% (95% CI, NE to NE) of patients achieved a complete response to treatment. The results of subgroup analyses were consistent with those of the FAS. The clinical experts consulted by CADTH indicated that the ORR and CRR results were clinically important and acceptable surrogates for survival outcomes (PFS and OS) based on their clinical experience in treating patients with r/r FL where extended survival is common, and therefore trials with mature survival follow-up are rare.

Survival outcomes were identified by CADTH with input from patient groups and clinicians as the most important efficacy outcomes to assess treatment effect in patients with r/r FL. Prolonged survival may be correlated with high response rates (e.g., ORR and CRR). In a meta-analysis evaluating the relationship between response rates and median PFS in patients with NHL (including FL, which accounted for 23% of the study population), strong correlation between response rates and PFS was found, the coefficient of determination (R2) was 0.78 for ORR versus median PFS and 0.74 for CRR versus median PFS. The results were similar in the subgroup of patients with r/r FL and treatment-naive FL.60 In another meta-analysis examining the correlation between response and survival outcomes, a moderate correlation was observed between CRR and median PFS in patients with FL (R2 = 0.69). In this study, the authors noted that since the median OS was usually not reached in clinical trials of NHL, none of the median OS-related correlation analysis results were evaluable due to the limited data.61 The clinical experts supported the use of surrogate outcomes by noting that, in their experience, patients who achieve complete remission after CAR T-cell therapy have better prognosis (e.g., more favourable survival) compared to those who do not respond well, but this is not always the case.

At the 36-month analysis time point in the ZUMA-5 trial, the survival probability for patients treated with axicabtagene ciloleucel was 75.5% (95% CI, 66.9% to 82.2%) and the median OS had not been reached. PFS was measured from the time of leukapheresis to the date of disease progression or death due to any cause. The proportion of patients who were progression free at the 36-month time point was 54.4% (95% CI, 44.2% to 63.5%), the median PFS was 40.2 months (95% CI, 28.9 months to NE). According to the clinical experts consulted by CADTH, the survival results from the ZUMA-5 trial are consistent with the expected clinical benefit; however, it must be noted that due to the slowly progressing nature of r/r FL, the OS and PFS data are immature and therefore the effect on long-term survival is uncertain. Results from a retrospective analysis conducted in a single centre in the US showed that median OS was 11.7 years, 8.8 years, and 5.3 years for patients who received second-line, third-line, and fourth-line treatments, respectively. In this study, recurrent uses of single-agent rituximab (9% to 31%), alkylator-based chemotherapy (22% to 26%), and radiotherapy (alone or radioimmunotherapy, 10% to 18%) were common in second- to sixth-line treatment therapy. Ten percent of the treated patients received SCT during their course of therapy (auto-SCT 6%; allo-SCT 4%). Investigational therapies (not specified, and unclear whether CAR T-cell products were used) ranged from 8% to 22% when second or later lines of therapy were required.62 Furthermore, when considering the long survival periods in r/r FL, it is important to take into account the impact of any beneficial therapies that could be introduced in the future and the impact that they may have on the long-term survival of patients who received axicabtagene ciloleucel in the ZUMA-5 trial.

HRQoL was identified by patient groups and clinicians as an important outcome. The ZUMA-5 trial did not include any HRQoL end points, and therefore the effects of axicabtagene ciloleucel on HRQoL in patients with r/r FL is unknown.

Given the single-arm design of the ZUMA-5 trial, there is no head-to-head evidence against standard of care for patients with r/r FL. A comparison of axicabtagene ciloleucel against an external standard of care control arm from the retrospective SCHOLAR-5 cohort found that axicabtagene ciloleucel is associated with improved OS and PFS. The interpretation of the comparative efficacy estimates is limited by the potential for selection bias when accepting patients into the ZUMA-5 clinical trial, and residual imbalances in important prognostic and effect-modifying patient characteristics, despite propensity score weighting.

It is also noted that tisagenlecleucel is another CAR T-cell therapy currently under review at CADTH for use in patients with r/r FL after 2 or more lines of therapy.63 No comparative evidence between axicabtagene and tisagenlecleucel was identified, and therefore the comparative efficacy is unknown.

Harms

At the 36-month time point, ||| of patients in the ZUMA-5 trial had reported at least 1 AE. The most common were pyrexia (|||), hypotension (|||||), headache (|||), and fatigue (|||||). SAEs were reported in ||| of patients, with the most common being pyrexia (|||||). It was also noted that given that axicabtagene ciloleucel is administered as a 1-time infusion, the ZUMA-5 trial follow-up is sufficient to characterize the safety profile. The clinical experts highlighted the adverse events of special interest, specifically CRS that occurred in ||| of patients (|| at grade ≥ 3) and neurologic events that occurred in ||| of patients (||| at grade ≥ 3). The clinical experts noted that it appears that patients have different rates of CRS and immune effector cell–associated neurotoxicity syndrome. The experts indicated that patients’ characteristics, such as performance status and comorbidities that might predict their ability to tolerate an episode of CRS or immune effector cell–associated neurotoxicity syndrome, might influence the choice of product. However, given the lack of head-to-head evidence or an indirect treatment comparison, the comparative safety profiles are unknown.

The SCHOLAR-5 comparison did not include safety as an end point, and therefore the safety profile of axicabtagene ciloleucel compared to standard of care is unknown. The clinical experts consulted by CADTH considered the safety profile of axicabtagene ciloleucel to be as expected given the mechanism of action and prior experience in other indications; therefore, side effects are expected to be manageable with proper monitoring.

Conclusion

Evidence from a single-arm study (ZUMA-5) suggests that treatment with axicabtagene ciloleucel affects clinically important tumour responses, including complete remission, in adult patients with r/r FL after 2 or more lines of systemic therapies. Due to the single-arm design of the trial and limited duration of follow-up, there is insufficient evidence to determine the magnitude of the effect of axicabtagene ciloleucel on OS and PFS. HRQoL outcomes were not included in the ZUMA-5 trial and therefore the impact of axicabtagene ciloleucel on patients’ HRQoL is unknown. The harms associated with the axicabtagene infusion are as expected given the mechanism of action and prior experience in other indications. The comparison of the ZUMA-5 trial to the retrospective SCHOLAR-5 external control was limited by heterogeneity across study designs and populations. Specifically, the inability to adjust for ECOG PS and double refractory status can introduce bias to the estimation procedure within the comparative populations. Generalizability to individuals who do not meet the ZUMA-5 trial criteria is also in question. Therefore, the magnitude of the comparative efficacy estimates for axicabtagene ciloleucel against standard of care in the Canadian setting is uncertain.

References

1.Lymphoma Canada. Understanding NHL: A patient's guide to Non-Hodgkin Lymphoma. 2023; https://z9a2f9c2.rocketcdn.me/wp-content/uploads/2020/05/2020430-NHL-Interior-WEB.pdf. Accessed 2023 May 4.

2.Canadian Cancer Society. What is Non-Hodgkin lymphoma? 2023; https://cancer.ca/en/cancer-information/cancer-types/non-hodgkin-lymphoma/what-is-non-hodgkin-lymphoma. Accessed 2023 May 4.

3.National Cancer Institute. Non-Hodgkin lymphoma treatment (PDQ®)–health professional version. 2023; https://www.cancer.gov/types/lymphoma/hp/adult-nhl-treatment-pdq#_1. Accessed 2023 May 4.

4.Hubel K, Ghielmini M, Ladetto M, Gopal AK. Controversies in the treatment of follicular lymphoma. Hemasphere. 2020;4(1):e317. PubMed

5.Jaffe ES. Follicular lymphomas: a tapestry of common and contrasting threads. Haematologica. 2013;98(8):1163-1165. PubMed

6.Le M, Ghazawi FM, Alakel A, et al. Incidence and mortality trends and geographic patterns of follicular lymphoma in Canada. Curr Oncol. 2019;26(4):e473-e481. PubMed

7.Pongas G, Cheson B. Recent Advances in the Management of Patients with Relapsed/Refractory Follicular Lymphoma. Blood Lymphat Cancer. 2021;11:55-66. PubMed

8.Ellison LF, Wilkins K. Canadian trends in cancer prevalence. Health Rep. 2012;23(1):7-16. PubMed

9.Canadian Cancer Society. Canadian cancer statistics. 2021; https://cdn.cancer.ca/-/media/files/research/cancer-statistics/2021-statistics/2021-pdf-en-final.pdf. Accessed 2022 Nov 30.

10.Horning SJ. Natural history of and therapy for the indolent non-Hodgkin's lymphomas. Semin Oncol. 1993;20(5 Suppl 5):75-88. PubMed

11.Horning SJ, Rosenberg SA. The natural history of initially untreated low-grade non-Hodgkin's lymphomas. NEJM. 1984;311(23):1471-1475. PubMed

12.Johnson PW, Rohatiner AZ, Whelan JS, et al. Patterns of survival in patients with recurrent follicular lymphoma: a 20-year study from a single center. J Clin Oncol. 1995;13(1):140-147. PubMed

13.Liu J, Stewart D, Fontaine A, et al. Characteristics and outcomes of patients with relapsed follicular lymphoma following retreatment with second-line rituximab-containing chemotherapy. Leuk Lymphoma. 2020;61(10):2492-2496. PubMed

14.Rivas-Delgado A, Magnano L, Moreno-Velazquez M, et al. Response duration and survival shorten after each relapse in patients with follicular lymphoma treated in the rituximab era. Br J Haematol. 2019;184:753-759. PubMed

15.Erblich T, Montoto S. Treating relapsed follicular lymphoma. Expert Rev Hematol. 2018;11(5):403-410. PubMed

16.Cancer Reasearch UK. Non-Hodgkin lymphoma- follicular lymphoma. 2020; https://www.cancerresearchuk.org/about-cancer/non-hodgkin-lymphoma/types/follicular-lymphoma#:~:text=Follicular%20lymphoma%20is%20a%20type,normally%20helps%20us%20fight%20infections. Accessed 2023 May 4.

17.Canadian Cancer Society. Survival statistics for non-Hodgkin lymphoma. 2023; https://cancer.ca/en/cancer-information/cancer-types/non-hodgkin-lymphoma/prognosis-and-survival/survival-statistics. Accessed 2023 May 4.

18.Clinical Study Report KTE-C19-105 (ZUMA-5): A phase 2 multicenter study of axicabtagene ciloleucel in subjects with relapsed/refractory indolent non-Hodgkin lymphoma. NCT03105336 [internal sponsor's report]. Santa Monica (CA): Kite Pharma, Inc; 2021.

19.A comparison of clinical outcomes from ZUMA-5 (axicabtagene ciloleucel) and the international SCHOLAR-5 External Control Cohort In Relapsed/Refractory Follicular Lymphoma (R/R FL): 36-month follow-up for patients with three or more prior lines of therapy and patients with two or more prior lines of therapy, addendum to modules 5.3.5.2 ZUMA-5 clinical study report. [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: axicabtagene ciloleucel, target of 2 × 106 chimeric antigen receptor (CAR)-positive viable T cells per kg body weight with a maximum of 2 × 108 CAR-positive viable T cells, IV. Mississauga (ON): Gilead Sciences Canada; 2023 Apr 13.

20.Armitage JO, Gascoyne RD, Lunning MA, Cavalli F. Non-Hodgkin lymphoma. Lancet. 2017;390(10091):298-310. PubMed

21.Canadian Cancer Society. Non-Hodgkin lymphoma statistics. 2022; https://cancer.ca/en/cancer-information/cancer-types/non-hodgkin-lymphoma/statistics. Accessed 2023 May 4.

22.Ye X, Mahmud S, Skrabek P, Lix L, Johnston JB. Long-term time trends in incidence, survival and mortality of lymphomas by subtype among adults in Manitoba, Canada: a population-based study using cancer registry data. BMJ Open. 2017;7(7):e015106. PubMed

23.Sarkozy C, Maurer MJ, Link BK, et al. Cause of Death in Follicular Lymphoma in the First Decade of the Rituximab Era: A Pooled Analysis of French and US Cohorts. J Clin Oncol. 2019;37(2):144-152. PubMed

24.Canadian Cancer Society. Follicular lymphoma. 2023; https://cancer.ca/en/cancer-information/cancer-types/non-hodgkin-lymphoma/what-is-non-hodgkin-lymphoma/follicular-lymphoma. Accessed 2023 May 4.

25.Montoto S, Davies AJ, Matthews J, et al. Risk and clinical implications of transformation of follicular lymphoma to diffuse large B-cell lymphoma. J Clin Oncol. 2007;25(17):2426-2433. PubMed

26.Kuruvilla J, Assouline S, Hodgson D, et al. A Canadian evidence-based guideline for the first-line treatment of follicular lymphoma: joint consensus of the Lymphoma Canada Scientific Advisory Board. Clin Lymphoma Myeloma Leuk. 2015;15(2):59-74. PubMed

27.Solal-Céligny P, Roy P, Colombat P, et al. Follicular Lymphoma International Prognostic Index. Blood. 2004;104(5):1258-1265. PubMed

28.Casulo C, Byrtek M, Dawson KL, et al. Early relapse of follicular lymphoma after rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone defines patients at high risk for death: An analysis from the National LymphoCare Study. J Clin Oncol. 2015;33(23):2516-2522. PubMed

29.UHN Princess Margaret Cancer Centre. Lymphoma: indolent lymphomas. (Clinical Practice Guidelines) 2020; https://www.uhn.ca/PrincessMargaret/Health_Professionals/Programs_Departments/Documents/CPG_Lymphoma_IndolentLymphomas.pdf. Accessed 2023 Jan 5.

30.B-Cell lymphomas. (NCCN Guidelines). Plymouth Meeting (PA): National Comprehensive Cancer Network; 2022 Jul 12: https://www.nccn.org/professionals/physician_gls/pdf/b-cell.pdf. Accessed 2022 Oct 4.

31.Follicular lymphoma. (NCCN Guidelines for Patients.). Plymouth Meeting (PA): National Comprehensive Cancer Network; 2022: https://www.nccn.org/patients/guidelines/content/PDF/nhl-follicular-patient.pdf. Accessed 2022 Oct 4.

32.López-Guillermo A, Caballero D, Canales M, Provencio M, Rueda A, Salar A. Clinical practice guidelines for first-line/after-relapse treatment of patients with follicular lymphoma. Leuk Lymphoma. 2011;52 Suppl 3:1-14. PubMed

33.Dreyling M, Ghielmini M, Rule S, et al. Newly diagnosed and relapsed follicular lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2021;32(3):298-308. PubMed

34.Alberta Health Services Guideline Resource Unit. Lymphoma. (Clinical Practice Guideline LYHE-002 V16.) 2021; https://www.albertahealthservices.ca/assets/info/hp/cancer/if-hp-cancer-guide-lyhe002-lymphoma.pdf. Accessed 2022 Oct 5.

35.Lymphoma, chronic leukemia, myeloma: Hodgkin lymphoma. (Cancer Management Manual). Vancouver (BC): BC Cancer Agency; 2013: http://www.bccancer.bc.ca/health-professionals/clinical-resources/cancer-management-manual/lymphoma-chronic-leukemia-myeloma/hodgkin-lymphoma#Treatment. Accessed 2022 Oct 5.

36.Lymphoma Canada. Follicular lymphoma guidelines. 2015; http://www.lymphoma.ca/medpath/#md:cid=1&md:viewname=dashboardTreeView&ca:viewname=CADefault&ca:id=404&car:viewname=CourseActivityResourcesEmpty&cat:viewname=CourseActivityTreeActivityByType. Accessed 2022 Oct 5.

37.Robinson SP, Canals C, Luang JJ, et al. The outcome of reduced intensity allogeneic stem cell transplantation and autologous stem cell transplantation when performed as a first transplant strategy in relapsed follicular lymphoma: an analysis from the Lymphoma Working Party of the EBMT. Bone Marrow Transplant. 2013;48(11):1409-1414. PubMed

38.Buske C, Hutchings M, Ladetto M, et al. ESMO Consensus Conference on malignant lymphoma: general perspectives and recommendations for the clinical management of the elderly patient with malignant lymphoma. Ann Oncol. 2018;29(3):544-562. PubMed

39.Klyuchnikov E, Bacher U, Woo Ahn K, et al. Long-term survival outcomes of reduced-intensity allogeneic or autologous transplantation in relapsed grade 3 follicular lymphoma. Bone Marrow Transplant. 2016;51(1):58-66. PubMed

40.Smith SM, Godfrey J, Ahn KW, et al. Autologous transplantation versus allogeneic transplantation in patients with follicular lymphoma experiencing early treatment failure. Cancer. 2018;124(12):2541-2551. PubMed

41.Sarkozy C, Trneny M, Xerri L, et al. Risk Factors and Outcomes for Patients With Follicular Lymphoma Who Had Histologic Transformation After Response to First-Line Immunochemotherapy in the PRIMA Trial. J Clin Oncol. 2016;34(22):2575-2582. PubMed

42.Teva-Lenalidomide (lenalidomide) 2.5 mg, 5 mg, 10 mg, 15 mg, 20 mg, and 25 mg capsules [product monograph]. Toronto (ON): Teva Canada Ltd; 2022 Sep 13.

43.Lenalidomide plus rituximab chemotherapy for relapsed or refractory indolent B-cell Non-Hodgkin lymphomas. (CADTH Health Technology Review). Can J Health Technol. Vol 12021: https://www.cadth.ca/sites/default/files/pdf/htis/2021/RC1392-R2-RR-NHL_Final.pdf. Accessed 2023 Oct 25.

44.Zydelig (idelalisib) 150 mg and 100 mg tablets [product monograph]. Mississauga (ON): Gilead Sciences Canada Inc; 2015 Feb 19, revised 2020 Aug 18.

45.CADTH Pan-Canadian Oncology Drug Review (pCODR) Expert Review Committee (pERC) final recommendation: Idelalisib (Zydelig - Gilead Sciences). Ottawa (ON): CADTH; 2016: https://www.cadth.ca/sites/default/files/pcodr/pcodr_idelalisib_zydelig_fl_fn_rec.pdf. Accessed 2022 Oct 31.

46.National Cancer Institute. CAR T cells: engineering patients' immune cells to treat their cancers. 2022; https://www.cancer.gov/about-cancer/treatment/research/car-t-cells. Accessed 2023 Apr 8.

47.Kymriah (tisagenlecleucel) cell suspension in infusion bag, 1.2 x 106 to 6.0 x 108 CAR-positive viable T cells for intravenous use [product monograph] Montreal (QC): Novartis Pharmaceuticals Canada; 2018, revised 2022: https://www.ask.novartispharma.ca/download.htm?res=kymriah_scrip_e.pdf&resTitleId=1520. Accessed 2022 Dec 19.

48.Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol. 2014;32(27):3059-3068. PubMed

49.Yescarta® (axicabtagene ciloleucel): cell suspension in patient-specific single infusion bag, target of 2 × 106 chimeric antigen receptor (CAR)-positive viable T cells per kg body weight with a maximum of 2 × 108 CAR-positive viable T cells, for intravenous infusion [product monograph]. Mississauga (ON): Gilead Sciences Canada, Inc.; 2021 Dec 15.

50.Jacobson C, Chavez J, Sehgal A, et al. Interim analysis of ZUMA-5: A phase 2 study of axicabtagene ciloleucel (Axi-cel) in patients with relapsed/refractory indolent non-Hodgkin lymphoma. Am J Hematol. 2020;95(SUPPL 1):S20-S21.

51.Jacobson C, Chavez JC, Sehgal AR, et al. Primary Analysis of Zuma-5: A Phase 2 Study of Axicabtagene Ciloleucel (Axi-cel) in Patients with Relapsed/Refractory (R/R) Indolent Non-Hodgkin Lymphoma (iNHL). Blood. 2020;136(Supplement 1):40-41.

52.Neelapu SS, Chavez JC, Sehgal AR, et al. Long-term follow-up analysis of zuma-5: A phase 2 study of axicabtagene ciloleucel (AXI-CEL) in patients with relapsed/refractory (R/R) indolent non-Hodgkin lymphoma (INHL). Blood. 2021;138(SUPPL 1):93.

53.Jacobson CA, Chavez JC, Sehgal AR, et al. Axicabtagene ciloleucel in relapsed or refractory indolent non-Hodgkin lymphoma (ZUMA-5): a single-arm, multicentre, phase 2 trial. Lancet Oncol. 2022;23(1):91-103. PubMed

54.Evans SR. Clinical trial structures. J Exp Stroke Transl Med. 2010;3(1):8-18. PubMed

55.Sun GW, Shook TL, Kay GL. Inappropriate use of bivariable analysis to screen risk factors for use in multivariable analysis. J Clin Epidemiol. 1996;49(8):907-916. PubMed

56.NCT05371093: Study of axicabtagene ciloleucel versus standard of care therapy in participants with relapsed/refractory follicular lymphoma (ZUMA-22). ClinicalTrials.gov. Bethesda (MD): U.S. National Library of Medicine; 2023: https://classic.clinicaltrials.gov/ct2/show/study/NCT05371093. Accessed 2023 Jul 14.

57.Dreyling M, Santoro A, Mollica L, et al. Phosphatidylinositol 3-kinase inhibition by copanlisib in relapsed or refractory indolent lymphoma. J Clin Oncol. 2017;35(35):3898-3905. PubMed

58.Sato T, Matsuyama Y. Marginal structural models as a tool for standardization. Epidemiology. 2003;14(6):680-686. PubMed

59.Clinical Evidence Template Yescarta (axicabtagene ciloleucel [internal sponsor's report]. Gilead Sciences Canada, Inc.; 2022 In: Drug Reimbursement Review sponsor submission: axicabtagene ciloleucel, target of 2 × 106 chimeric antigen receptor (CAR)-positive viable T cells per kg body weight with a maximum of 2 × 108 CAR-positive viable T cells, IV. Mississauga (ON): Gilead Sciences Canada; 2023 Apr 13.

60.Mangal N, Salem AH, Li M, Menon R, Freise KJ. Relationship between response rates and median progression-free survival in non-Hodgkin's lymphoma: A meta-analysis of published clinical trials. Hematol Oncol. 2018;36(1):37-43. PubMed

61.Zhu R, Lu D, Chu YW, et al. Assessment of correlation between early and late efficacy endpoints to identify potential surrogacy relationships in non-Hodgkin lymphoma: A literature-based meta-analysis of 108 phase II and phase III studies. AAPS J. 2017;19(3):669-681. PubMed

62.Batlevi CL, Sha F, Alperovich A, et al. Follicular lymphoma in the modern era: survival, treatment outcomes, and identification of high-risk subgroups. Blood Cancer J. 2020;10(7):74. PubMed

63.Tisagenlecleucel for relapsed or refractory follicular lymphoma. (CADTH Health Technology Review). Ottawa (ON): CADTH; 2023: https://www.cadth.ca/tisagenlecleucel. Accessed 2023 Jul 14.

Pharmacoeconomic Review

Abbreviations

AE

adverse event

allo-SCT

allogeneic stem cell transplant

auto-SCT

autologous stem cell transplant

axi-cel

axicabtagene ciloleucel

CAR

chimeric antigen receptor

CRS

cytokine release syndrome

FL

follicular lymphoma

ICER

incremental cost-effectiveness ratio

ICU

intensive care unit

KM

Kaplan-Meier

OCCI

Ontario Case Costing Initiative

OS

overall survival

PD

progressed disease

PFS

progression-free survival

QALY

quality-adjusted life-year

R2

lenalidomide plus rituximab

r/r

relapsed or refractory

SCT

stem cell transplant

SOC

standard of care

WTP

willingness to pay

Executive Summary

The executive summary comprises 2 tables (Table 1 and Table 2) and a conclusion.

Table 1: Submitted for Review

Item

Description

Drug product

Axicabtagene ciloleucel (Yescarta), cell suspension of CAR-positive viable T-cells, for IV infusion.

Submitted price

Axicabtagene ciloleucel, cell suspension of 2 × 106 CAR T cells/kg body weight, to a maximum of 2 × 108 CAR T cells: $485,021 per 1-time infusion.

Indication

Adult patients with relapsed or refractory grade (r/r) grade 1, 2, or 3a follicular lymphoma (FL) after two or more lines of systemic therapy.

Health Canada approval status

NOC/c

Health Canada review pathway

Standard review

NOC date

September 28, 2022

Reimbursement request

As per indication

Sponsor

Gilead Sciences Canada Inc.

Submission history

Previously reviewed: Yes

Indication: Treatment of adult patients with DLBCL or HGBL that is refractory to first-line chemoimmunotherapy or that relapses within 12 months of first-line chemoimmunotherapy.

Recommendation date: February 3, 2023

Recommendation: Reimburse with clinical criteria and conditions

Indication: Treatment of adult patients with r/r LBCL after 2 or more lines of systemic therapy.

Recommendation date: August 15, 2019

Recommendation: Recommended with clinical criteria and conditionsa

CAR = chimeric antigen receptor; DLBCL = diffuse large B-cell lymphoma; FL = follicular lymphoma; HGBL = high-grade B-cell lymphoma; LBCL = large B-cell lymphoma; NOC = Notice of Compliance; NOC/c = Notice of Compliance with conditions; r/r = relapsed or refractory.

aThis review of axicabtagene ciloleucel went through the interim review process for CAR T-cell therapies, in which recommendations were issued by the CADTH Health Technology Expert Review Panel.

Table 2: Summary of Economic Evaluation

Component

Description

Type of economic evaluation

Cost-utility analysis

Partitioned survival model

Target population

Adult patients with r/r grade 1, 2, or 3a FL after ≥ 2 lines of systemic therapy

Treatment

Axicabtagene ciloleucel

Comparator

SOC is composed of chemotherapy (50%), SCT (12%), idelalisib (5%), and clinical trials (33%).

Chemotherapy includes 6 different regimens:

  • rituximab plus bendamustine

  • CHOP

  • CVP

  • obinutuzumab plus bendamustine

  • GDP

  • R-CVP

Perspective

Canadian publicly funded health care payer

Outcomes

QALYs, life-years

Time horizon

Lifetime (50 years)

Key data source

  • Axi-cel: single-arm, phase II ZUMA-5 trial (36-month data cut-off: March 31, 2022)

  • SOC: SCHOLAR-5 retrospective cohort study (patients who initiated third or higher line of therapy on or after July 2014)

  • Comparative efficacy data were informed from the indirect treatment comparison of SCHOLAR-5 and ZUMA-5 studies through propensity score weighting on prespecified prognostic factors using standardized mortality ratios.

Submitted results

ICER = $115,543 per QALY gained compared with SOC (incremental costs = $505,565; incremental QALYs = 4.38).

Key limitations

  • The sponsor implemented a cure model that assumed that 40% of patients receiving axi-cel who remain progression-free for 5 years would be considered clinically cured. CADTH noted that it is premature to determine the fraction and time point upon which patients would achieve long-term remission given that (1) follow-up in the ZUMA-5 trial is limited; (2) long remissions are common among patients with FL; and (3) permanence of CAR T-cell treatment efficacy is uncertain.

  • The magnitude and durability of the survival benefit with axi-cel is highly uncertain in the absence of more robust head-to-head evidence. Clinical experts indicated that it is plausible that the OS due to axi-cel treatment could converge with that of SOC within the model’s lifetime horizon, that is, for axi-cel’s treatment effect to wane within patients’ lifetimes.

  • The parametric distribution selected by the sponsor to model long-term OS for patients receiving SOC in the economic model underestimated both the KM estimates informed by the sponsor-submitted SCHOLAR-5 retrospective cohort study and the median OS derived from real-world evidence.

  • The sponsor failed to consider the upfront costs associated with assessment of CAR T-cell therapy eligibility. In addition, the sponsor underestimated the pretreatment costs of leukapheresis for patients receiving CAR T-cell therapy.

  • The sponsor assigned different utility estimates to be accrued by patients with PD according to subsequent treatment status. Clinical experts indicated that quality of life is not expected to differ between those who are on subsequent treatment and those who are off subsequent treatment.

  • The sponsor omitted the R2 regimen from the analysis despite evidence that the therapy is used off-label in current Canadian clinical practice.

CADTH reanalysis results

  • CADTH reanalyses were derived by making changes to the following model parameters: using standard parametric models based on KM data from the ZUMA-5 trial to extrapolate the OS and PFS of axi-cel for the entire duration of the model; using alternative parametric models to extrapolate the OS of SOC and axi-cel; and including a CAR T-cell therapy eligibility assessment cost and updating the pretreatment cost associated with leukapheresis. Given the magnitude of uncertainty of the effect of axi-cel treatment on OS, its comparative efficacy against SOC, and the durability of such a benefit, CADTH conducted separate analyses involving different parametric assumptions for OS.

  • In CADTH reanalysis A, the OS for axi-cel was modelled using the exponential distribution (assuming treatment effect for 15.3 years postinfusion before waning). Axi-cel was associated with an ICER of $544,875 per QALY gained compared to SOC (incremental costs: $505,223; incremental QALYs: 0.93). A price reduction of 95% would be required for axi-cel to be cost-effective at a WTP threshold of $50,000 per QALY gained.

  • In CADTH reanalysis B, the OS for axi-cel was modelled using the log-normal distribution (assuming treatment effect would be maintained for the entire time horizon of the model). Axi-cel was associated with an ICER of $243,879 per QALY gained compared to SOC (incremental costs: $505,885; incremental QALYs: 2.07). Under this reanalysis, a price reduction of 82% would be required for axi-cel to be cost-effective at a WTP threshold of $50,000 per QALY gained.

axi-cel = axicabtagene ciloleucel; CAR = chimeric antigen receptor; CHOP = cyclophosphamide, doxorubicin, vincristine, and prednisone; CVP = cyclophosphamide, vincristine, and prednisone; FL = follicular lymphoma; GDP = gemcitabine, dexamethasone, and cisplatin; ICER = incremental cost-effectiveness ratio; KM = Kaplan-Meier; OS = overall survival; PD = progressed disease; PFS = progression-free survival; QALY = quality-adjusted life-year; R2 = lenalidomide plus rituximab; R-CVP = rituximab plus cyclophosphamide, vincristine, and prednisone; r/r = relapsed or refractory; SOC = standard of care; SCT = stem cell transplant; WTP = willingness to pay.

Conclusions

Evidence from the ZUMA-5 single-arm trial suggests that treatment with axi-cel may be associated with clinically important tumour responses, including complete remission, in adult patients with relapsed or refractory (r/r) FL after 2 or more lines of systemic therapies. However, there is insufficient evidence — due to the single-arm design of the trial as well as limited follow-up duration — to determine the effects of axi-cel on overall survival (OS) and progression-free survival (PFS). In addition, the CADTH clinical assessment identified limitations with the sponsor’s comparison of the ZUMA-5 trial to the SCHOLAR-5 trial, which substantially restricted the ability to interpret the treatment effects of axi-cel relative to that of SOC. Overall, the Clinical Review concluded that there is a high degree of uncertainty around the comparative treatment effects of axi-cel relative to SOC.

Given the magnitude of uncertainty to do with the effect of axi-cel treatment on OS, its comparative efficacy against SOC, and the durability of such a benefit, CADTH was unable to derive a robust base-case estimate of cost-effectiveness. Moreover, given the duration of the ZUMA-5 trial (36 months) in contrast to the model’s time horizon (50 years), it is important to note that the majority of the quality-adjusted life-year (QALY) benefit was derived from the time period beyond which there are observed trial data (i.e., extrapolated period). To address this, CADTH conducted separate reanalyses involving different parametric assumptions of treatment effect waning for OS: (A) using the exponential distribution that assumes 15.3 years of treatment effect postinfusion before waning; and (B) using the log-normal distribution that assumes sustained treatment effect for the entire time horizon of the model.

In CADTH reanalysis A (assuming 15.3 years of treatment effect postinfusion before waning), axi-cel was associated with 0.93 incremental QALYs gained and additional costs of $505,223 relative to SOC, resulting in an incremental cost-effectiveness ratio (ICER) of $544,875 per QALY gained. In CADTH reanalysis B (assuming that treatment effect would be maintained for the entire time horizon of the model), axi-cel was associated with 2.07 incremental QALYs gained and additional costs of $505,885 relative to SOC, resulting in an ICER of $243,879 per QALY gained. The estimated ICERs were higher than the sponsor’s base-case value, driven primarily by removing the cure assumption. In line with clinical expert advice, these reanalyses achieved more plausible OS curves in the absence of robust long-term evidence, while still conferring a benefit with axi-cel. CADTH noted that both reanalyses assume life expectancy increases for patients receiving axi-cel relative to current SOC (2.68 and 1.10 years of life gained in reanalysis A and B, respectively). However, the true impact of axi-cel on OS relative to SOC remains uncertain in the absence of evidence from randomized studies. The CADTH reanalyses assume that the impacts of residual confounding that could influence the nonrandomized comparison of the ZUMA-5 and SCHOLAR-5 studies are limited and that their findings could be replicated in real-world clinical practice. Both assumptions are highly uncertain. Given the available evidence, the estimates presented within the CADTH reanalyses may represent the upper bounds of the incremental gains that may be realized from this therapy.

Assuming OS and PFS outcomes from the nonrandomized comparison of the ZUMA-5 and SCHOLAR-5 studies can be replicated in real-world settings and extensions in lifespan occur relative to current SOC, a price reduction of between 82% and 95% would be required for axi-cel to be cost-effective at a willingness-to-pay (WTP) threshold of $50,000 per QALY gained. This would reduce the 1-time price of axi-cel from $485,021 to $86,334 (an 82% price reduction) or $26,676 (a 95% price reduction). This range reflects the uncertainty around long-term survival extrapolation as analyzed in CADTH reanalyses A and B.

Stakeholder Input Relevant to the Economic Review

This section is a summary of the feedback received from the patient groups, registered clinicians, and drug plans that participated in the CADTH review process.

One patient group, Lymphoma Canada, provided input through data collected via an online survey. The survey, conducted from April 2022 to April 2023, included 143 patients with lymphoma (of whom 34% were diagnosed with follicular lymphoma [FL] and 86% lived in Canada). Of note, 3 respondents reported having experience with axicabtagene ciloleucel (axi-cel). The most important outcomes, according to the respondents, included delaying disease progression and achieving long-term remission, with the ultimate objective of improving survival; reducing side effects from treatments; preserving independence to minimize the burden on caregivers; and maintaining quality of life. Based on survey responses, 49% of patients underwent a period of “watchful waiting” before starting treatment. The majority of the patients surveyed (43%) had received 1 line of therapy, while 20% had received 2 and 16% had received 3, where chemoimmunotherapy was the most commonly prescribed treatment. The majority of patients surveyed received 2 regimens: rituximab plus cyclophosphamide, hydroxydaunorubicin (doxorubicin), oncovin (vincristine), and prednisone; and cyclophosphamide, vincristine, and prednisone. Important side effects of chemotherapy included fatigue, low activity level, and hair loss. The patients emphasized the need for therapies that can be administered at a hospital located near home to minimize travel time and burden on caregivers, as well as improve quality of life by keeping patients close to their support systems. Patients who had experience with axi-cel accessed this therapy via enrolment in a clinical trial. They reported travelling out of province to receive treatment and being away from home for up to 3 months to do so. Patients who received axi-cel experienced side effects that included cytokine release syndrome (CRS), neutropenia, febrile neutropenia, thrombocytopenia, constipation, and swelling.

Registered clinician input was received from Ontario Health-Cancer Care Ontario Hematology Cancer Drug Advisory Committee. According to the clinicians, the current pathway of care for patients with r/r FL after 2 or more lines of systemic therapy is chemoimmunotherapy and autologous stem cell transplant (auto-SCT). The clinicians noted that in select patients for whom the treatment goal is mostly palliative; allogeneic stem cell transplant (allo-SCT) and radiotherapy may also be considered. The clinicians suggested chemoimmunotherapy would likely be less efficacious among re-treated patients, thus pointing to the need for additional therapy options. The clinicians suggested that axi-cel may shift the current treatment paradigm by replacing chemoimmunotherapy in third line, but will likely not replace auto-SCT among eligible patients. Although it is uncertain whether axi-cel could replace auto-SCT in third line, the clinicians noted the potential for axi-cel to be prescribed in advance of auto-SCT among patients who are refractory to chemotherapy. Furthermore, it was noted that axi-cel should only be considered in relatively fit patients without significant comorbidities. Patients with uncontrolled infections, severe organ dysfunction, and poor performance status should be excluded. Clinician input further noted that axi-cel should be considered in select patients who had received prior CD19-directed therapy or allo-SCT, despite their exclusion from the pivotal trial.

Participating drug plans noted concerns that the existing capacity may not be able to meet the anticipated demand in Canada. Given the requirement for specialized and accredited treatment centres where the therapy can be administered, access may require interprovincial travel and, without full coverage of interprovincial reimbursement, may impact equitable access across Canada. Finally, drug plans queried whether, and how, potential manufacturing delays may impact the clinical effectiveness of axi-cel.

Several of these concerns were addressed in the sponsor’s model:

In addition, CADTH addressed some of these concerns as follows:

CADTH was unable to address the following concerns raised from stakeholder input:

Economic Review

The current review is for axi-cel (Yescarta) for adult patients with r/r grade 1, 2, or 3a FL after 2 or more lines of systemic therapy.

Economic Evaluation

Summary of Sponsor’s Economic Evaluation

Overview

The sponsor submitted a cost-utility analysis of axi-cel compared with SOC. Aligned with Health Canada’s indicated population, the modelled population comprised adult patients with r/r grade 1, 2, or 3a FL after 2 or more lines of systemic therapy.1

Axi-cel is a CD19-directed genetically modified autologous T-cell immunotherapy individually prepared from a patient’s peripheral blood mononuclear cells.1 It is available as a cell suspension for infusion containing a target dose of 2 × 106 chimeric antigen receptor (CAR) T-cells/kg body weight (range, 1 × 106 cells/kg to 2.4 × 106 cells/kg), to a maximum of 2 × 108 CAR T cells for patients weighing 100 kg or more.2 It is provided as a single-dose, 1-time infusion. The sponsor’s submitted price for axi-cel is $485,021 per infusion,1 not including costs associated with pretreatment (i.e., leukapheresis, bridging chemotherapy, and lymphodepleting chemotherapy), hospitalization related to inpatient administration, and postinfusion stay in an intensive care unit (ICU) stay.

SOC, the comparator for this analysis, encompassed a basket of therapies commonly used in Canadian clinical practice. SOC was composed of 50% chemotherapy, 12% SCT, 5% phosphoinositide 3-kinase inhibitors (i.e., idelalisib), and 33% investigational therapies offered though clinical trials.1 The treatment cost associated with chemotherapy was estimated as a weighted average of 6 chemotherapy regimens: rituximab plus bendamustine; cyclophosphamide, hydroxydaunorubicin, vincristine, and prednisone; cyclophosphamide, vincristine, and prednisone; obinutuzumab plus bendamustine; gemcitabine, dexamethasone, and cisplatin; rituximab plus cyclophosphamide, vincristine, and prednisone.1

Based on clinician-informed proportions of patients on each chemotherapy regimen and average number of cycles per regimen, the sponsor estimated the total weighted drug cost of chemotherapy to be $15,650.1 The sponsor estimated the cost of auto-SCT and allo-SCT to be $70,434 and $91,992, respectively, which incorporated the cost of stem cell transplant (SCT) procedures (including high-dose chemotherapy, stem cell harvest, and infusion), as well as the cost of inpatient stay associated with administration.1 The SOC composite cost was estimated as a weighted average of the drug acquisition and administration costs associated with chemotherapy, SCT, phosphoinositide 3-kinase inhibitors, and investigational therapies accessed through clinical trials ($32,073).1 Vial-sharing was not incorporated by the sponsor.

The clinical outcomes modelled were PFS and OS.1 The economic outcomes of interest were QALYs and life-years. The economic evaluation was conducted over a lifetime time horizon (50 years), from the perspective of the Canadian public health care payer. Costs and outcomes were discounted at 1.5% per annum.1

Model Structure

The sponsor used a partitioned survival model to capture all costs and outcomes associated with axi-cel and SOC. The model included 3 health states: progression-free, progressed disease (PD), and death, with transitions between health states occurred on a monthly cycle length (Figure 1). The proportion of patients in progression-free, PD, and death states was estimated over time based on OS and PFS curves, which were informed by the ZUMA-5 single-arm trial, as well as the SCHOLAR-5 retrospective cohort study. The proportion of patients with PD (i.e., post-progression state) was estimated as the difference between the proportion of living patients (estimated from the OS curve) and the proportion of progression-free patients (estimated from the PFS curve). PFS was defined as the time from the date of enrolment to the date of first documented progression or death due to any cause. Patients began in the progression-free health state and, over time, could transition to either the PD health state or the death state. Patients in the PD health state could remain either in this health state or transition to the death state (i.e., patients could not return to the progression-free health state).

Model Inputs

Baseline patient characteristics were derived from the ZUMA-5 trial, a phase II, single-arm, multicentre trial investigating the efficacy and safety of axi-cel among patients with r/r grade 1, 2, or 3a FL (n = 127).1 The typical patient in the modelled cohort, which the sponsor assumed reflected the patient population in Canada, was aged | | years and weighed | | kg, and was more likely to be male (59%).1 These characteristics were those of the patient population enrolled in the ZUMA-5 trial and were used to inform the drug dosage regimens and the age- and sex-specific distribution of the general population mortality risk.1

Clinical efficacy parameters used to characterize axi-cel and SOC, including OS and PFS, were derived from various data sources. For axi-cel, inputs were based on the 36-month follow-up analysis of the ZUMA-5 single-arm trial (data cut-off: March 31, 2022). For SOC, inputs were informed by the SCHOLAR-5 study, a multicentre, international, observational, retrospective study that constructed a historical control cohort of patients with r/r FL treated with 2 or more prior lines of usual therapies in routine practice (n = 128). The SCHOLAR-5 cohort was derived from 3 international cohorts: IQVIA, Vanderbilt, and DELTA. The IQVIA and Vanderbilt cohorts were created from electronic medical records, while the DELTA cohort represented patients from the DELTA clinical trial who proceeded to receive therapy subsequent to idelalisib. The DELTA cohort was added to increase statistical power for OS. In the absence of head-to-head evidence, the sponsor conducted an indirect treatment comparison to assess the relative efficacy of axi-cel versus SOC therapies. Propensity score methods, specifically standardized mortality ratio weighting, were applied to account for the imbalance of confounders between the ZUMA-5 trial and the SCHOLAR-5 external control cohort.3 Specifically, the OS and PFS for axi-cel and SOC were estimated using a weighted Kaplan-Meier (KM) estimator, whereby patients from the SCHOLAR-5 external cohort were weighted to be comparable with the ZUMA-5 trial population across baseline covariates using propensity score matching.3

Survival data from the indirect comparison of the ZUMA-5 and SCHOLAR-5 study results were extrapolated to derive the long-term survival estimates of OS and PFS informing the economic model.1 Standard parametric models were fit independently to the individual patient data from the ZUMA-5 and SCHOLAR-5 studies. Under the assumption that axi-cel was curative, the sponsor derived piecewise cure models to estimate OS and PFS, which accounted for the proportion of patients receiving axi-cel who may experience long-term survival. The piecewise cure models estimated a likelihood of cure (i.e., “statistically cured” fraction), wherein the survival outcomes for the “cured” group, relative to the “uncured” group, which does not achieve long-term remission, are assumed to be better. Cured patients experience a slightly worse survival than the general population (hazard ratio = 1.09),4 while uncured patients experience poorer survival outcomes relative to cancer-free individuals of the same age and sex. In the submitted base case, 40% of patients receiving axi-cel were expected to achieve long-term remission (i.e., the cure fraction), with long-term survival applied 5 years from the start of the model (i.e., the cure time point). Hence, in the sponsor’s piecewise cure model, (1) standard parametric extrapolations for OS and PFS were applied until the 5-year cure time point; and (2) OS and PFS were calculated thereafter as the weighted product of background survival (for the cured population) and cancer-specific survival (for the uncured fraction). For patients receiving axi-cel, the sponsor used a piecewise cure model based on the exponential distribution to model OS, and a piecewise cure model based on the log-normal distribution to model PFS. Given that the sponsor did not extend the assumption of cure to patients receiving SOC therapies, the sponsor selected the standard exponential distribution to model both the OS and PFS of SOC. Survival distributions were selected based on Akaike information criterion and Bayesian information criterion, as well as visual inspection.1

As health-related quality of life data were not collected in the ZUMA-5 trial, health state utility values were obtained from the literature. Utility values were derived from 2 sources that reported health state utilities for indolent non-Hodgkin lymphoma using the EQ-5D results of 222 patients in the UK. The sponsor sourced utility values of 0.805 for the progression-free state, 0.620 for the PD on-treatment state, and 0.736 for the PD off-treatment state. Utility values were adjusted using age-related utility decrements based on an algorithm developed by Ara et al. (2010)5 to account for the natural decline in quality of life associated with age. The model incorporated disutilities associated with AEs categorized as grade 3 or greater in any of the treatments considered.1 Disutilities were applied as a 1-time decrement to the first model cycle; thus, it was assumed that AEs would have no further impact on costs beyond the initial hospitalization period. For axi-cel, AE rates were derived from those occurring in at least 5% of patients in the ZUMA-5 trial (36-month data cut-off);6 while for SOC, rates were calculated as a weighted average of AEs reported in clinical trials of SOC regimens.7-9

Costs captured in the model included pretreatment cost (i.e., drug acquisition and drug administration associated with pretreatment), treatment cost (i.e., drug acquisition, drug administration, and hospitalization associated with treatment), follow-up medical costs before progression (i.e., physician visits, PET/CT scans, and laboratory tests), subsequent treatment costs in fourth line, follow-up medical costs in postprogression, AE management costs, and terminal care costs.1 Drug acquisition costs for axi-cel were based on the sponsor’s submitted price.1 The dosing modelled for axi-cel is consistent with that described in the overview section. Drug acquisition costs were sourced from the Ontario Drug Benefit Formulary,10 with dosing schedules based on the chemotherapy regimen monographs from Ontario Health-Cancer Care Ontario.11

Prior to infusion, patients receiving axi-cel underwent leukapheresis, bridging chemotherapy, and conditioning chemotherapy. Given that data from the ZUMA-5 trial were used to determine the proportion of patients that would receive each phase of treatment, weighted costs were modelled for axi-cel. Pretreatment costs were applied in the first cycle of the model. The cost of leukapheresis ($2,688) was applied to all patients receiving axi-cel.12 The cost of bridging chemotherapy ($25) was estimated as the weighted 1-cycle cost of the following therapies: 60% dexamethasone ($3); 10% radiotherapy ($157); and 30% gemcitabine, dexamethasone, and cisplatin regimen ($652). This cost was applied to 3.1% of patients receiving axi-cel.1 Moreover, all patients receiving axi-cel were assumed to receive fludarabine plus cyclophosphamide administered daily for 3 days as conditioning chemotherapy ($2,300).1 Hospitalization and ICU inputs for axi-cel were estimated based on the average length of stay (| || || |) and proportion of patients in ICU (||||||) observed in the ZUMA trial as these were assumed to be reflective of Canadian clinical practice. Costs typically associated with the ongoing monitoring were obtained from the Canadian Institute for Health Information’s Patient Cost Estimator for an inpatient hospitalization for malignant lymphoma.13

For patients receiving SOC, treatment costs included chemotherapy (50%), SCT (12%), idelalisib (5%), and clinical trials (33%), based on a review of Canadian clinical practice guidelines and clinical expert opinion. The treatment costs associated with the SOC basket of therapies were as previously described. All regimens were assumed to be given in an outpatient setting, except for SCT. The cost of outpatient administration was based on the cost of complex chemotherapy administration from the Ontario Schedule of Benefits for Physician Services,14 and the cost associated with chair time was estimated based on the CCO regimen monographs.11 SCT procedure costs were obtained from the Interprovincial Health Insurance Agreements Coordinating Committee,15 and the Ontario Case Costing Initiative (OCCI).16

The total weighted costs of subsequent therapy differed by prior treatment (i.e., those who received CAR T cells versus SOC as third-line therapy) and were applied as a one-off cost in the first model cycle after progression. The weighted cost associated with subsequent therapy among patients receiving axi-cel and SOC in third line was estimated to be $14,714 and $6,871, respectively. Treatment monitoring costs and health care resource use costs were sourced from the Ontario Ministry of Health Schedule of Benefits for Laboratory and Physician Services.17 Treatment-emergent AE costs were estimated based on the data from the OCCI and applied as a 1-time cost in the first model cycle. All patients who transitioned to the death state were assumed to incur terminal care costs ($68,703) in the last cycle before death, based on the average cost for patients with terminal lymphoma in the Ontario Cancer Registry.19

Summary of Sponsor’s Economic Evaluation Results

The sponsor conducted the base case via a probabilistic sensitivity analysis with 1,000 simulations. The deterministic and probabilistic results were similar. The probabilistic findings are presented below.1

Base-Case Results

Compared with SOC, axi-cel was associated with an incremental QALY gain of 4.38 and an incremental cost of $505,565, resulting in an ICER of $115,543 per QALY (Table 3).1 Notably, the sponsor’s analysis predicted that axi-cel was associated with a longer duration of life than SOC (i.e., incremental life-years: 5.83).

Given the duration of the ZUMA-5 trial (i.e., 36 months) in contrast to the model’s time horizon (i.e., 50 years), it is important to note that the majority of the QALY and life-year benefit (96% and 97%, respectively) realized by patients receiving axi-cel was derived from the time period beyond which there are observed trial data (i.e., extrapolated period). Most of the QALYs gained by patients receiving axi-cel were realized in the progression-free state (78%), whereas patients receiving SOC realized most of their QALY gains in the PD state (78%). The key cost driver among patients receiving axi-cel was the cost of drug acquisition, accounting for 81% of the total cost incurred by patients. The main cost driver among patients receiving SOC was end-of-life care, which accounted for 55% of the total estimated cost.

Axi-cel was not cost-effective at a WTP threshold of $50,000 per QALY in any of the iterations when compared to SOC. The sponsor’s submitted analysis is based on the publicly available prices for all drug treatments. Additional results from the sponsor’s submitted economic evaluation base case are presented in Appendix 3.

Table 3: Summary of the Sponsor’s Economic Evaluation Results

Drug

Total costs ($)

Incremental costs ($)

Total QALYs

Incremental QALYs

ICER vs. SOC ($/QALY)

SOC

$111,964

Reference

4.90

Reference

Reference

Axi-cel

$617,528

$505,565

9.28

4.38

$115,543

axi-cel = axicabtagene ciloleucel; ICER = incremental cost-effectiveness ratio; QALY = quality-adjusted life-year; SOC = standard of care; vs. = versus.

Source: Sponsor’s pharmacoeconomic submission.1

Sensitivity and Scenario Analysis Results

The sponsor assessed several model parameters and assumptions in probabilistic scenario analyses. These included: applying a shorter model time horizon; using another analysis set from the ZUMA-5 trial; applying different health state utility values; assessing the impact of different cure fractions; applying alternative parametric distributions to model the OS and PFS of axi-cel and SOC; and aligning the axi-cel re-treatment proportion with that observed in the ZUMA-5 trial. The parameters with the greatest influence were other assumptions regarding efficacy, particularly selection of OS extrapolations and cure fraction, as well as shorter time horizons. When applying a 10% cure fraction for axi-cel, the ICER increased to $168,794 per QALY gained. Moreover, when applying a 10-year time horizon, the ICER increased to $375,698 per QALY gained. All other scenarios resulted in ICERs ranging between $92,912 and $162,936 per QALY gained.1

CADTH Appraisal of the Sponsor’s Economic Evaluation

CADTH identified several key limitations to the sponsor’s analysis that have notable implications on the economic analysis:

Additionally, the following key assumptions were made by the sponsor and have been appraised by CADTH (See Table 5).

Table 4: Key Assumptions of the Submitted Economic Evaluation

Sponsor’s key assumption

CADTH comment

AEs qualified as grade ≥ 3 were incorporated in the model with an associated cost and disutility, because they were observed in ≥ 5% of patients in the ZUMA-5 trial. These were only applied to patients during the first cycle of the model, as it was assumed that AEs would have no further impact on costs and quality of life beyond the initial hospitalization period.

Not appropriate. This approach would be appropriate if all treatment-emergent AEs grade ≥ 3 occurred within the first month of treatment with axi-cel. However, patients in the ZUMA-5 trial experienced AEs beyond the first month of therapy. This is unlikely to be a key source of uncertainty.

Given that all eligible lines of treatment for each patient were included in the SCHOLAR-5 analysis set, the sponsor opted to randomly select the index line of therapy for patients with ≥ 2 prior lines of therapy. As such, the index treatment patterns for the SCHOLAR-5 cohort are not the same as the basket of therapies that patients receive as SOC in the third-line setting in the economic model.

Reasonable. CADTH noted that there is a misalignment between the third-line basket of therapies used by the SCHOLAR-5 patients (whose outcomes are used to derive the OS and PFS for SOC) and the third-line SOC therapy usage assumed by the sponsor in the model. While it was possible to alter the distribution of SOC therapies in line with Canadian clinical practice, doing so only impacted the cost of SOC and not the underlying OS estimates for SOC. Given that there are no established clinical practice guidelines for treating r/r FL in Canada (particularly after second-line therapy), the SOC options tend to be variable and dependent on what patients may have failed on previously. However, the SOC therapies included in the economic model were among the most prevalent in the SCHOLAR-5 analysis set. CADTH further noted that although simplifying assumptions were made, in general, these assumptions are reasonable. This is unlikely to be a key source of uncertainty.

AE = adverse event; axi-cel = axicabtagene ciloleucel; FL = follicular lymphoma; OS = overall survival; PFS = progression-free survival; r/r = relapsed or refractory; SOC = standard of care.

CADTH Reanalyses of the Economic Evaluation

Base-Case Results

CADTH reanalyses were derived by making changes to model parameter values and assumptions, in consultation with clinical experts. The following changes were made to address several limitations within the economic model: using standard parametric models based on KM data from the ZUMA-5 trial to extrapolate the OS and PFS of axi-cel for the entire duration of the model; using alternative parametric models to extrapolate the OS of SOC and axi-cel; and including a CAR T-cell therapy eligibility assessment cost and updating the pretreatment cost associated with apheresis. However, given the magnitude of uncertainty to do with the comparative clinical efficacy and durability of treatment effect of axi-cel relative to SOC, CADTH was unable to derive a robust base-case estimate of cost-effectiveness. CADTH conducted separate reanalyses involving different parametric assumptions of treatment effect for axi-cel. In reanalysis A, the treatment effect of axi-cel on OS was modelled using the exponential distribution; in reanalysis B, the treatment effect of axi-cel on OS was modelled using the log-normal distribution. These changes are summarized in Table 5.

Table 5: CADTH Revisions to the Submitted Economic Evaluation

Stepped analysis

Sponsor’s value or assumption

CADTH value or assumption

Changes to derive the CADTH base case

1. Assumption of cure of patients with FL is inappropriate

Piecewise cure models selected to extrapolate the OS and PFS for axi-cel

Standard parametric models selected to extrapolate the OS and PFS for axi-cel.

2. Impact of SOC on long-term OS is underestimated

OS for SOC was modelled using the exponential distribution.

OS for SOC was modelled using the log-logistic distribution.

3. Exclusion of CAR T-cell–related costs

  • Excluded CAR T-cell therapy eligibility assessment cost.

  • Cost associated with apheresis ($2,688) is underestimated.

  • Included CAR T-cell therapy eligibility assessment cost ($3,000).

  • Updated cost associated with apheresis ($5,426).

4. Impact of axi-cel on long-term OS is uncertaina

OS for axi-cel was modelled using a piecewise cure model based on the exponential distribution.

OS for axi-cel was modelled using the exponential distribution.

5. Impact of axi-cel on long-term OS is uncertaina

OS for axi-cel was modelled using a piecewise cure model based on the exponential distribution.

OS for axi-cel was modelled using the log-normal distribution.

CADTH reanalysis A

Reanalyses 1 + 2 + 3 + 4

CADTH reanalysis B

Reanalyses 1 + 2 + 3 + 5

axi-cel = axicabtagene ciloleucel; CAR = chimeric antigen receptor; OS = overall survival; PFS = progression-free survival; SOC = standard of care.

aCADTH reanalyses 4 and 5 (which change the axi-cel parametric distribution of OS to exponential and log-normal, respectively), require that reanalyses 1 and 2 be performed concurrently, that is, the axi-cel extrapolation method must be standard parametric (reanalysis 1) and the OS for SOC modelled using the log-logistic distribution (reanalysis 2).

Results from CADTH reanalyses A and B were generally aligned: axi-cel is not cost-effective at a $50,000 WTP threshold compared to SOC. In CADTH reanalysis A (assuming 15.3 years of treatment effect postinfusion before waning), axi-cel was associated with an ICER of $544,875 per QALY gained compared to SOC (incremental costs: $505,223; incremental QALYs: 0.93). In CADTH reanalysis B (assuming that treatment effect would be maintained for the entire model time horizon), axi-cel was associated with an ICER of $243,879 per QALY gained compared to SOC (incremental costs: $505,885; incremental QALYs: 2.07). The probability that axi-cel was cost-effective at a WTP threshold of $50,000 per QALY was 0% in both reanalyses A and B.

The estimated ICERs were higher than the sponsor’s base-case value, driven primarily by the use of standard parametric models based on KM data from the ZUMA-5 trial to extrapolate the OS and PFS of axi-cel (i.e., rejecting the assumption of a statistically cured fraction). In line with clinical expert advice, these reanalyses achieved more plausible OS curves in the absence of long-term evidence, while still conferring a survival benefit with axi-cel. In both reanalyses, most incremental QALYs were due to improvements in life-years. Furthermore, 76% (reanalysis A) and 89% (reanalysis B) of QALYs gained by patients receiving axi-cel were derived from the extrapolated period in which there are no observed trial data. The majority of the total cost among patients receiving axi-cel was associated with drug acquisition costs (78%), while the key cost driver among patients receiving SOC was related to terminal care costs (51%).

CADTH reanalyses are based on the publicly available prices for all drug treatments. Full results are available in Appendix 4.

Table 6: Summary of the Stepped Analysis of the CADTH Reanalysis Results (Deterministic)

Stepped analysis

Drug

Total costs ($)

Total QALYs

ICER ($/QALY)

Sponsor’s base case (deterministic)

SOC

$111,947

4.87

Reference

Axi-cel

$617,582

9.26

$115,232

CADTH reanalysis 1: cure assumption

SOC

$111,947

4.87

Reference

Axi-cel

$615,331

7.50

$190,994

CADTH reanalysis 2: SOC OS (log-logistic)

SOC

$113,102

6.96

Reference

Axi-cel

$617,582

9.26

$220,155

CADTH reanalysis 3: CAR T-cell costs

SOC

$114,947

4.87

Reference

Axi-cel

$623,320

9.26

$115,855

CADTH reanalysis 4: axi-cel OS (exponential)

SOC

$113,102

6.96

Reference

Axi-cel

$615,594

7.90

$536,574

CADTH reanalysis 5: axi-cel OS (log-normal)

SOC

$113,102

6.96

Reference

Axi-cel

$616,285

9.11

$235,080

CADTH reanalysis A: (1 + 2 + 3 + 4)

SOC

$116,102

6.96

Reference

Axi-cel

$621,332

7.90

$539,497

CADTH reanalysis B: (1 + 2 + 3 + 5)

SOC

$116,102

6.96

Reference

Axi-cel

$622,022

9.11

$236,359

CADTH reanalysis A: probabilistic

SOC

$116,105

6.97

Reference

Axi-cel

$621,329

7.90

$544,875

CADTH reanalysis B: probabilistic

SOC

$116,105

6.97

Reference

Axi-cel

$621,991

9.05

$243,879

axi-cel = axicabtagene ciloleucel; CAR = chimeric antigen receptor; ICER = incremental cost-effectiveness ratio; OS = overall survival; QALY = quality-adjusted life-year; SOC = standard of care.

Scenario Analysis Results

CADTH undertook price reduction analyses based on the sponsor’s results and CADTH’s reanalyses. Results of CADTH reanalysis A suggested a price reduction of 95% would be required to achieve cost-effectiveness of axi-cel relative to SOC at a $50,000 per QALY threshold. In CADTH reanalysis B, a price reduction of 82% would be required (Table 7).

Table 7: CADTH Price Reduction Analyses

Analysis

ICERs for axi-cel vs. SOC

Price reduction

Sponsor base case

CADTH reanalysis A

CADTH reanalysis B

No price reduction

$115,231

$539,487

$236,356

10%

$104,178

$487,696

$213,697

20%

$93,125

$435,906

$191,038

30%

$82,071

$384,115

$168,378

40%

$71,018

$332,324

$145,719

50%

$59,965

$280,533

$123,060

60%

$48,911

$228,742

$100,401

70%

$37,858

$176,951

$77,741

80%

$26,805

$125,161

$55,082

90%

$15,751

$73,370

$32,423

100%

$4,698

$21,579

$9,764

axi-cel = axicabtagene ciloleucel; ICER = incremental cost-effectiveness ratio; SOC = standard of care; vs. = versus.

CADTH undertook 1 scenario analysis on each of the CADTH reanalyses. In line with clinical expert advice, CADTH assumed that for patients with r/r FL who have PD, quality of life would not differ according to subsequent treatment status. Specifically, CADTH assumed utility equivalence between patients who are on and off subsequent treatment (0.736). In reanalysis A, the ICER increased to $564,126 (incremental costs: $505,223; incremental QALYs: 0.90). In reanalysis B, the ICER increased to $246,379 (incremental costs: $505,885; incremental QALYs: 2.05).

The results of this analysis are presented in Table 14.

Issues for Consideration

Overall Conclusions

Evidence from the ZUMA-5 single-arm trial suggests that treatment with axi-cel may be associated with clinically important tumour responses, including complete remission, in adult patients with r/r FL after 2 or more lines of systemic therapies. However, there is insufficient evidence — due to the single-arm design of the trial as well as limited follow-up duration — to determine the effects of axi-cel on OS and PFS. The CADTH clinical assessment identified limitations with the sponsor’s comparison of the ZUMA-5 trial to the SCHOLAR-5 study, which substantially restricted the ability to interpret the relative treatment effects of axi-cel and SOC. Overall, the CADTH Clinical Review concluded that there is a high degree of uncertainty around the treatment effects of axi-cel relative to SOC.

Given the magnitude of uncertainty to do with the effect of axi-cel treatment on OS, axi-cel’s efficacy compared to SOC, and the durability of any benefit, CADTH was unable to derive a robust base-case estimate of cost-effectiveness. Moreover, given the duration of the ZUMA-5 trial (36 months) in contrast to the model’s time horizon (50 years), it is important to note that the majority of the QALY benefit realized by patients receiving axi-cel was derived from the time period beyond which there are observed trial data (i.e., extrapolated period). To address this, CADTH conducted separate reanalyses involving different parametric assumptions of treatment effect: in reanalysis A, the OS of patients receiving axi-cel was modelled using the exponential distribution (thus, assuming 15.3 years of treatment effect postinfusion before waning); and in reanalysis B, the OS of patients receiving axi-cel was modelled using the log-normal distribution (thus, assuming that the treatment effect of axi-cel, relative to SOC, would be maintained for the entire time horizon of the model). In addition, the following changes were made consistently across reanalyses A and B to address limitations within the economic model: using standard parametric models based on KM data from the ZUMA-5 trial to extrapolate the OS and PFS of axi-cel for the entire duration of the model; using alternative parametric models to extrapolate the OS of SOC and axi-cel; and including a CAR T-cell eligibility assessment cost and updating the pretreatment cost associated with apheresis.

Results from CADTH reanalyses A and B were generally aligned: axi-cel is not cost-effective at a $50,000 WTP threshold compared to SOC. In CADTH reanalysis A (assuming 15.3 years of treatment effect postinfusion before waning), axi-cel was associated with an ICER of $544,875 per QALY gained compared to SOC (incremental costs: $505,223; incremental QALYs: 0.93). In CADTH reanalysis B (assuming that treatment effect would be maintained for the entire time horizon of the model), axi-cel was associated with an ICER of $243,879 per QALY gained compared to SOC (incremental costs: $505,885; incremental QALYs: 2.07). The estimated ICERs were higher than the sponsor’s base-case value, driven primarily by the use of standard parametric models based on KM data from the ZUMA-5 trial to extrapolate the OS and PFS of axi-cel (i.e., rejecting the assumption of a statistically cured fraction). In line with clinical expert advice, these reanalyses achieved more plausible OS curves in the absence of robust long-term evidence, while still conferring a benefit with axi-cel. CADTH noted that both reanalyses assume life expectancy increases for patients receiving axi-cel relative to current SOC (2.68 and 1.10 years of life gained in reanalysis A and B, respectively). However, the true impact of axi-cel on OS relative to SOC remains uncertain in the absence of evidence from randomized trials. The CADTH reanalyses assume that the impacts of residual confounding that could influence the nonrandomized comparison of the ZUMA-5 and SCHOLAR-5 studies are limited and that their findings could be replicated in real-world clinical practice. Both assumptions are highly uncertain. Hence, given the available evidence, the estimates presented in the CADTH reanalyses likely represent the upper bounds of the incremental gains that may be realized from this therapy.

A price reduction of 95% or 82% would be required for axi-cel to be cost-effective at a WTP threshold of $50,000 per QALY gained, conditional on axi-cel’s long-term impact on OS relative to SOC. This would mean a reduction in the 1-time price of axi-cel from $485,021 to $86,334 (an 82% price reduction) or $26,676 (a 95% price reduction). This range reflects the uncertainty around long-term survival extrapolation as analyzed in CADTH reanalyses A and B. Finally, CADTH undertook 1 scenario analysis on each of the CADTH reanalyses. In line with clinical expert advice, CADTH assumed that among patients with r/r FL who have PD, quality of life would not differ according to subsequent treatment status. In reanalysis A, the ICER increased to $564,126 (incremental costs: $505,223; incremental QALYs: 0.90). In reanalysis B, the ICER increased to $246,379 (incremental costs: $505,885; incremental QALYs: 2.05).

References

1.Pharmacoeconomic evaluation [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: axicabtagene ciloleucel, target of 2 × 106 chimeric antigen receptor (CAR)-positive viable T cells per kg body weight with a maximum of 2 × 108 CAR-positive viable T cells, IV. Mississauga (ON): Gilead Sciences Canada; 2023 Apr 13.

2.Yescarta (axicabtagene ciloleucel): Cell suspension in patient-specific single infusion bag, target of 2 × 106 chimeric antigen receptor (CAR)-positive viable T cells per kg body weight with a maximum of 2 × 108 CAR-positive viable T cells, for intravenous infusion [product monograph]. Mississauga (ON): Gilead Sciences Canada; 2019 Feb 13.

3.A comparison of clinical outcomes from ZUMA-5 (axicabtagene ciloleucel) and the international SCHOLAR-5 external control cohort in relapsed/refractory follicular lymphoma (R/R FL): 36-month follow-up for patients with three or more prior lines of therapy and patients with two or more prior lines of therapy, addendum to modules 5.3.5.2 ZUMA-5 clinical study report. [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: axicabtagene ciloleucel, target of 2 × 106 chimeric antigen receptor (CAR)-positive viable T cells per kg body weight with a maximum of 2 × 108 CAR-positive viable T cells, IV. Mississauga (ON): Gilead Sciences Canada; 2023 Apr 13.

4.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):1066-1073. PubMed

5.Ara R, Brazier JE. Populating an economic model with health state utility values: moving toward better practice. Value Health. 2010;13(5):509-518. PubMed

6.Kite Pharma. Common Technical Document section 2.7.4: Summary of clinical safety [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: axicabtagene ciloleucel, target of 2 × 106 chimeric antigen receptor (CAR)-positive viable T cells per kg body weight with a maximum of 2 × 108 CAR-positive viable T cells, IV. Mississauga (ON): Gilead Sciences Canada; 2023 Apr 13.: Gilead; 2021.

7.Marcus R, Davies A, Ando K, et al. Obinutuzumab for the first-line treatment of follicular lymphoma. N Engl J Med. 2017;377(14):1331-1344. PubMed

8.Marcus R, Imrie K, Solal-Celigny P, et al. Phase III study of R-CVP compared with cyclophosphamide, vincristine, and prednisone alone in patients with previously untreated advanced follicular lymphoma. J Clin Oncol. 2008;26(28):4579-4586. PubMed

9.Hiddemann W, Kneba M, Dreyling M, et al. Frontline therapy with rituximab added to the combination of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) significantly improves the outcome for patients with advanced-stage follicular lymphoma compared with therapy with CHOP alone: results of a prospective randomized study of the German Low-Grade Lymphoma Study Group. Blood. 2005;106(12):3725-3732. PubMed

10.Ontario Ministry of Health and Long-Term Care. Ontario drug benefit formulary/comparative drug index. 2022; https://www.formulary.health.gov.on.ca/formulary/. Accessed 2023 Oct 26.

11.Cancer Care Ontario. Drug Formulary. GDP - Provider Monograph. 2019; https://www.cancercareontario.ca/en/drugformulary/regimens/monograph/46166. Accessed 2022 Aug 15.

12.Ellis K. Cost-effectiveness of chimeric antigen receptor T-cell therapy for treating large B-cell lymphoma patients in Canada [master's thesis]. Waterloo (ON): University of Waterloo; 2019: https://uwspace.uwaterloo.ca/bitstream/handle/10012/15515/Ellis_Kristina.pdf. Accessed 2023 Oct 26.

13.Patient cost estimator. Ottawa (ON): Canadian Institute for Health Information; 2022: https://www.cihi.ca/en/patient-cost-estimator. Accessed 2023 Oct 26.

14.Schedule of benefits for physician services under the Health Insurance Act: (January 25, 2022 (effective July 1, 2022)). Toronto (ON): Ontario Ministry of Health; 2022: https://www.health.gov.on.ca/en/pro/programs/ohip/sob/physserv/sob_master.pdf. Accessed 2023 Oct 26.

15.Interprovincial Health Insurance Agreements Coordinating Committee (IHIACC). Final - interprovincial billing rates for designated high cost transplants effective for discharges on or after April 1, 2022. Organ transplants. Toronto (ON): Ontario Ministry of Health, Ontario Ministry of Long-Term Care; 2022: https://files.ontario.ca/moh-2023-2024-rates-high-cost-procedures-en-2023-03-24.pdf. Accessed 2022 Oct 18.

16.Kearns B, Pandor A, Stevenson M, et al. Cabazitaxel for hormone-relapsed metastatic prostate cancer previously treated with a docetaxel-containing regimen: An evidence review group perspective of a NICE single technology appraisal. Pharmacoeconomics. 2017;35(4):415-424. PubMed

17.Trapani D, Tay-Teo K, Tesch ME, et al. Implications of oncology trial design and uncertainties in efficacy-safety data on health technology assessments. Curr Oncol. 2022;29(8):5774-5791. PubMed

18.Ambulatory care 2017/2018 by procedure. In: Ontario Case Costing Initiative (OCCI) [dataset]. Ottawa (ON): Ontario Ministry of Health and Long-Term Care; 2017: https://data.ontario.ca/dataset/ontario-case-costing-initiative-occi Accessed 2022 Dec.

19.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. PubMed

20.Chong EA, Ruella M, Schuster SJ. Five-year outcomes for refractory B-cell lymphomas with CAR T-cell therapy. N Engl J Med. 2021;384(7):673-674. PubMed

21.Zinzi A, Gaio M, Liguori V, et al. Late relapse after CAR-T cell therapy for adult patients with hematologic malignancies: a definite evidence from systematic review and meta-analysis on individual data. Pharmacol Res. 2023;190:106742. PubMed

22.Clinical Study Report KTE-C19-105 (ZUMA-5): A phase 2 multicenter study of axicabtagene ciloleucel in subjects with relapsed/refractory indolent non-Hodgkin lymphoma: 36-month follow-up analysis [internal sponsor's report]. Santa Monica (CA): Kite Pharma, Inc; 2022.

23.Everest L, Blommaert S, Chu RW, Chan KKW, Parmar A. Parametric survival extrapolation of early survival data in economic analyses: A comparison of projected versus observed updated survival. Value Health. 2022;25(4):622-629. PubMed

24.Ye X, Mahmud S, Skrabek P, Lix L, Johnston JB. Long-term time trends in incidence, survival and mortality of lymphomas by subtype among adults in Manitoba, Canada: a population-based study using cancer registry data. BMJ Open. 2017;7(7):e015106. PubMed

25.Latimer NR. Survival analysis for economic evaluations alongside clinical trials--extrapolation with patient-level data: inconsistencies, limitations, and a practical guide. Med Decis Making. 2013;33(6):743-754. PubMed

26.Batlevi CL, Sha F, Alperovich A, et al. Follicular lymphoma in the modern era: survival, treatment outcomes, and identification of high-risk subgroups. Blood Cancer J. 2020;10(7):74. PubMed

27.Pettengell R, Donatti C, Poynton C, et al. The impact of follicular lymphoma on health-related quality of life. Ann Oncol. 2008;19:570-576. PubMed

28.Wild D, Walker M, Pettengell R, Lewis G. PCN62 utility elicitation in patients with follicular lymphoma. Value Health. 2006;9(6):A294.

29.Gilead. CADTH Reimbursement Review: Reimbursement Status for Comparators [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: axicabtagene ciloleucel, target of 2 × 106 chimeric antigen receptor (CAR)-positive viable T cells per kg body weight with a maximum of 2 × 108 CAR-positive viable T cells, IV. Mississauga (ON): Gilead Sciences Canada; 2023 Apr 13.

30.Gilead. CADTH Reimbursement Review Implementation Planning for Cell or Gene Therapy. [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: axicabtagene ciloleucel, target of 2 × 106 chimeric antigen receptor (CAR)-positive viable T cells per kg body weight with a maximum of 2 × 108 CAR-positive viable T cells, IV. Mississauga (ON): Gilead Sciences Canada; 2023 Apr 13.

31.Li SZJ, Hallard D, Fontaine MP, Grzegorczyl-Manuel I, L. B. Abstract 27 (poster): Manufacturing commercial axicabtagene ciloleucel for Canadian patients: a retrospective analysis [conference abstract]. Cell Therapy Transplant 2022 Annual Conference (CTTC 2022); 2022 June 15-18; Niagara Falls, Ontario. Curr Oncol. 2022;29:10005-10006.

32.Keating SJ, Gu T, Jun MP, McBride A. Health care resource utilization and total costs of care among patients with diffuse large B cell lymphoma treated with chimeric antigen receptor t cell therapy in the United States. Transplant Cell Ther. 2022;28(7):404.e401-404.e406. PubMed

33.Axicabtagene ciloleucel for diffuse large B-cell lymphoma: economic report. (CADTH optimal use report vol. 9, no. 1d). Ottawa (ON): CADTH; 2019: https://www.cadth.ca/sites/default/files/pdf/car-t/ct0002-axicabtagene-ciloleucel-economic-report-redacted.pdf. Accessed 2022 Aug 18.

34.CADTH Reimbursement review: lisocabtagene maraleucel (Breyanzi) for the treatment of relapsed or refractory large B-cell lymphoma. Can J Health Technol. 2022;2(10). https://www.cadth.ca/sites/default/files/DRR/2022/PG0258-Breyanzi.pdf. Accessed 2022 Aug 20.

35.Verma AA, Pai M, Saha S, et al. Managing drug shortages during a pandemic: tocilizumab and COVID-19. CMAJ. 2021;193(21):E771-E776. PubMed

36.DeltaPA. [Ottawa (ON)]: IQVIA; 2022: https://www.iqvia.com/. Accessed 2023 Oct 26.

37.BC Cancer. CLIC-01 clinical trial: building BC Cancer’s capabilities in cutting-edge immunotherapy. 2023 Jan 12; http://www.bccancer.bc.ca/about/news-stories/stories/clic-01-clinical-trial-building-bc-cancer%E2%80%99s-capabilities-in-cutting-edge-immunotherapy. Accessed 2023 Apr 11.

38.PrRinvoq® (upadacitinib): 15 mg, 30 mg extended-release oral tablets [product monograph]. St-Laurent (QC): AbbVie; 2021 Sep 30: https://www.abbvie.ca/content/dam/abbvie-dotcom/ca/en/documents/products/RINVOQ_PM_EN.pdf. Accessed 2022 Oct 1.

39.Hübel K, Ghielmini M, Ladetto M, Gopal AK. Controversies in the treatment of follicular lymphoma. Hemasphere. 2020;4(1):e317. PubMed

40.Canadian Cancer Statistics Advisory Committee. Canadian cancer statistics 2021. Toronto (ON): Canadian Cancer Society. In collaboration with Statistics Canada and the Public Health Agency of Canada; 2021: https://cdn.cancer.ca/-/media/files/research/cancer-statistics/2021-statistics/2021-pdf-en-final.pdf. Accessed 2023 Oct 26.

41.Table: 17-10-0057-01 Projected population, by projection scenario, age and sex, as of July 1 (x 1,000). Ottawa (ON): Statistics Canada; 2022: https://www150.statcan.gc.ca/t1/tbl1/en/tv.action?pid=1710005701. Accessed 2023 Oct 26.

42.Ontario Case Costing Initiative (OCCI) [dataset]. Toronto (ON): Ontario Health and Long-Term Care; 2017: https://data.ontario.ca/dataset/ontario-case-costing-initiative-occi. Accessed 2023 Oct 27.

Appendix 1: Cost Comparison Table

Note that this appendix has not been copy-edited.

The comparators presented in the following table have been deemed to be appropriate based on feedback from clinical expert(s) and drug plans. Comparators may be recommended (appropriate) practice or actual practice. Existing Product Listing Agreements are not reflected in the table and as such, the table may not represent the actual costs to public drug plans.

Table 8: CADTH Cost Comparison Table for r/r Grade 1, 2, or 3a FL After 2 or More Lines of Systemic Therapy (Gene Therapy)

Treatment

Strength / concentration

Form

Price ($)

Recommended dosage

Cost per cycle or 1-time use ($)

Cost per

28 days ($)

CAR T-cell therapy

Axicabtagene ciloleucel (Yescarta)

Refer to dosage

Suspension for IV infusion

485,021.0000a

Target of 2 × 106 anti-CD19 CAR T cells/kg body weight (range, 1 × 106 to 2.4 × 106 cells/kg) to a maximum of 2 × 108 anti-CD19 CAR T cells (1-time infusion)

485,021

NA

CAR = chimeric antigen receptor; FL = follicular lymphoma; NA = not applicable; r/r = relapsed or refractory.

Table 9: CADTH Cost Comparison Table for r/r Grade 1, 2, or 3a FL After 2 or More Lines of Systemic Therapy (Chemotherapy, SCT)

Treatment

Strength / concentration

Form

Price ($)

Recommended dosage

Cost per cycle or 1-time use ($)

Cost per

28 days ($)

Chemotherapy

BR

Bendamustine (generic)

5 mg/mL

25 mg vial

100 mg vial

with powder for solution

296.8800

1,062.5000

28-day cycles:

90 mg/m2 days 1 and 2c

3,906

3,906

Rituximab (biosimilar)

10 mg/mL

100 mg vial for IV infusion

297.0000b

28-day cycles:

375 mg/m2 on day 1c

2,079

2,079

BR regimen cost (21-day cycle)

5,985

5,985

CHOP

Cyclophos­phamide (Procytox)

20 mg/mL

500 mg vial

1,000 mg vial

2,000 mg vial

for IV infusion

93.1400

168.8300

310.6000

21-day cycles:

750 mg/m2 IV on day 1c

262

349

Doxorubicin (generic)

2 mg/mL

10 mg vial

50 mg vial

for IV infusion

50.0000

250.0000

21-day cycles:

50 mg/m2 IV on Day 1c

455

607

Vincristine (generic)

1 mg/mL

1 mg vial

2 mg vial

5 mg vial

for IV infusion

30.6000

62.0000

153.0000

21-day cycles:

1.4 mg/m2 IV (max 2 mg) on day 1c

62

83

Prednisone (generic)

50 mg

Tablet

0.1735

21-day cycles:

100 mg orally on days 1 to 5c

2

2

CHOP regimen cost (21-day cycle)

781

1,041

CVP

Cyclophos­phamide (Procytox)

20 mg/mL

500 mg vial

1,000 mg vial

2000 mg vial

for IV infusion

93.1400

168.8300

310.6000

21-day cycles:

750 mg/m2 IV on day 1c

262

349

Vincristine (generic)

1 mg/mL

1 mg vial

2 mg vial

5 mg vial

for IV infusion

30.6000

62.0000

153.0000

21-day cycles:

1.4 mg/m2 IV (max 2 mg) on day 1c

62

83

Prednisone (generic)

50 mg

Tablet

0.1735

21-day cycles:

100 mg PO on days 1 to 5c

2

2

CVP regimen cost (21-day cycle)

326

434

GB

Obinutuzumab

10 mg/mL

100 mg vial

for IV infusion

297.0000b

28-day cycles:

1,000 mg IV on days 1, 8, and 15 in cycle 1, and on day 1 on each cycle thereafterc

Induction:

15,827

Maintenance:

5,276

Induction:

15,827

Maintenance:

5,276

Bendamustine (generic)

5 mg/mL

25 mg vial

100 mg vial

with powder for solution

296.8800

1,062.5000

28-day cycles:

90 mg/m2 days 1 and 2c

3,906

3,906

GB induction regimen cost (21-day cycle)

19,733

19,733

GB maintenance regimen cost (21-day cycle)

9,182

9,182

GDP

Gemcitabine (generic)

40 mg/mL

1,000 mg vial

2,000 mg vial

with lyophilized powder for infusion

270.0000

540.0000

21-day cycles:

1,000 mg/m2 days 1 and 8c

1,080

1,440

Dexamethasone (generic)

4 mg

Tablet

0.3046

21-day cycles:

40 mg days 1 to 4c

12

16

Cisplatin (generic)

1 mg/mL

50 mg vial

100 mg vial

with solution for injection

135.0000

270.0000

21-day cycles:

75 mg/m2 on day 1c

405

540

GDP regimen cost (21-day cycle)

1,497

1,996

R-CVP

Rituximab (biosimilar)

10 mg/mL

100 mg vial

for IV infusion

297.0000b

21-day cycles:

375 mg/m2 on day 1c

2,079

2,772

Cyclophos­phamide (Procytox)

20 mg/mL

500 mg vial

1,000 mg vial

2,000 mg vial

for IV infusion

93.1400

168.8300

310.6000

21-day cycles:

750 mg/m2 IV on day 1c

262

349

Vincristine (generic)

1 mg/mL

1 mg vial

2 mg vial

5 mg vial

for IV infusion

30.6000

62.0000

153.0000

21-day cycles:

1.4 mg/m2 IV (max 2 mg) on day 1c

62

83

Prednisone (generic)

50 mg

Tablet

0.1735

21-day cycles:

100 mg orally dailyc

7

10

R-CVP regimen cost (21-day cycle)

2,410

3,214

PI3K inhibitor

Idelalisib (Zydelig)

100 mg

150 mg

Tablet

85.35

85.35

150 mg twice daily

NA

4,780

SCT

Autologous SCT (< 72 hours)

Adult autologous stem cell transplant (< 72 hours); includes all facility costs including inpatient and diagnostic costs

36,645d per transplant

NA

Autologous SCT (> 72 hours)

Adult autologous stem cell transplant (> 72 hours); includes all facility costs including inpatient and diagnostic costs

77,956e per transplant

NA

Allogeneic SCT (non-MUD patients)

Adult allogeneic stem cell transplant; includes all facility costs including inpatient and diagnostic costs; excludes MUD patients

179,392d per transplant

NA

Allogeneic SCT (MUD patients)

Adult allogeneic stem cell transplant; includes all facility costs including inpatient and diagnostic costs; MUD patients

216,542d per transplant

NA

CHOP = cyclophosphamide, doxorubicin, vincristine, and prednisone; CVP = cyclophosphamide, vincristine, and prednisone; FL = follicular lymphoma; GDP = gemcitabine, dexamethasone, and cisplatin; MUD = matched unrelated donor; PI3K = phosphoinositide 3 kinase; R2 = lenalidomide plus rituximab; R-CVP = rituximab plus cyclophosphamide, vincristine, and prednisone; SCT = stem cell transplant.

Note: All prices are wholesale from IQVIA Delta PA (accessed May 2023), unless otherwise indicated, and do not include dispensing fees. Calculations assume a patient body weight of 75 kg and a body surface area of 1.8 m2.

aSponsor’s submitted price.1

bOntario Drug Benefit Formulary or Exceptional Access Program list price10 (accessed May 2023).

cCancer Care Ontario Formulary: Regimens database.38

dInterprovincial Billing Rates for Designated High Cost Transplants Effective for Discharges on or after April 1, 2022.15 The cost includes all facility costs associated with a single transplant episode including inpatient and diagnostic costs.|

eInterprovincial Billing Rates for Designated High Cost Transplants Effective for Discharges on or after April 1, 2022.15 The cost includes all facility costs associated with a single transplant episode including inpatient and diagnostic costs, with a maximum length of stay of 16 days.

Appendix 2: Submission Quality

Note that this appendix has not been copy-edited.

Table 10: Submission Quality

Description

Yes or no

Comments

Population is relevant, with no critical intervention missing, and no relevant outcome missing.

No

The sponsor excluded the R2 regimen (lenalidomide + rituximab) from the base-case analysis, despite the fact that the therapy is used off-label for the treatment of adult patients with r/r grade 1, 2, or 3a FL after ≥ 2 lines of systemic therapy in Canada.

Model has been adequately programmed and has sufficient face validity.

No

The sponsor’s model was not thoroughly debugged. For instance, CADTH remarks that when selecting the generalized gamma parametric distribution to extrapolate the OS for axi-cel within a prespecified dropdown list, the probabilistic analysis could not be properly conducted as it would yield calculation errors across several iterations, which complicated the validation process.

Model structure is adequate for decision problem.

No

The PSM further introduces structural assumptions about the relationship between PFS and OS (i.e., non–mutually exclusive curves), which is potentially problematic since they are likely dependent outcomes. Clinical expert opinion suggested that survival is linked to the occurrence of progressive disease and thus the transition probability to death should vary for patients within the progression-free state compared to those in the progressive disease state.

Data incorporation into the model has been done adequately (e.g., parameters for probabilistic analysis).

Yes

No comment.

Parameter and structural uncertainty were adequately assessed; analyses were adequate to inform the decision problem.

Yes

No comment.

The submission was well organized and complete; the information was easy to locate (clear and transparent reporting; technical documentation available in enough details).

Yes

No comment.

FL = follicular lymphoma; OS = overall survival; PFS = progression-free survival; PSM = partitioned survival model; R2 = lenalidomide + rituximab.

Appendix 3: Additional Information on the Submitted Economic Evaluation

Note that this appendix has not been copy-edited.

Figure 1: Model Structure

Figure 1 presents the sponsor-submitted partitioned survival model structure composed of 3 health states: progression-free survival, progressed disease, and dead.

Tx = treatment.

Source: Sponsor’s pharmacoeconomic submission.1

Detailed Results of the Sponsor’s Base Case

Table 11: Disaggregated Summary of the Sponsor’s Base Case

Parameter

Axi-cel

SOC

Incremental

Discounted LYs

Total

12.39

6.54

5.85

Pre-progression

9.69

1.37

8.33

Post-progression

2.70

5.18

–2.48

Discounted QALYs

Total

9.26

4.87

4.39

Pre-progression

7.32

1.09

6.23

Post-progression

1.97

3.78

–1.81

AE Disutility

–0.03

0.00

–0.03

Discounted costs ($)

Total

$617,582

$111,947

$505,635

Pre-progression

$544,097

$34,950

$509,146

Drug costs

$487,347

$28,765

$458,581

Administration costs

$36,134

$3,308

$32,826

HCRU costs

$20,616

$1,998

$18,618

AE costs

$0

$879

–$879

Post-progression

$17,451

$14,983

$2,468

Drug costs

$12,592

$6,731

$5,861

Administration costs

$1,015

$875

$140

HCRU costs

$3,844

$7,377

–$3,533

Terminal care

$56,034

$62,013

–$5,979

ICER ($/QALY)

$115,232

AE = adverse event; axi-cel = axicabtagene ciloleucel; HCRU = health care resource use; ICER = incremental cost-effectiveness ratio; LY = life-year; QALY = quality-adjusted life-year; SOC = standard of care.

Appendix 4: Additional Details on the CADTH Reanalyses and Sensitivity Analyses of the Economic Evaluation

Note that this appendix has not been copy-edited.

Detailed Results of CADTH Base Case

Figure 2: OS for SOC and Axi-Cel in CADTH Reanalyses A and B

Figure 2 outlines OS extrapolations used to derive the CADTH reanalyses A and B for SOC and axi-cel.

Axi-cel; axicabtagene ciloleucel; OS = overall survival; SOC = standard of care.

Source: Sponsor’s pharmacoeconomic submission.1

Table 12: Disaggregated Summary of CADTH’s Reanalysis A

Parameter

Axi-cel

SOC

Incremental

Discounted LYs

Total

10.53

9.42

1.12

Pre-progression

7.03

1.37

5.66

Post-progression

3.51

8.05

–4.54

Discounted QALYs

Total

7.90

6.96

0.94

Pre-progression

5.37

1.09

4.28

Post-progression

2.56

5.87

–3.31

AE Disutility

–0.03

0.00

–0.03

Discounted costs ($)

Total

$621,332

$116,102

$505,230

Pre-progression

$544,162

$37,950

$506,212

Drug costs

$487,347

$28,765

$458,581

Administration costs

$41,872

$6,308

$35,564

HCRU costs

$14,944

$1,998

$12,946

AE costs

$0

$879

–$879

Post-progression

$19,237

$19,079

$158

Drug costs

$13,176

$6,731

$6,445

Administration costs

$1,062

$875

$187

HCRU costs

$4,999

$11,473

–$6,473

Terminal care

$57,932

$59,073

–$1,141

ICER ($/QALY)

$539,497

AE = adverse event; axi-cel = axicabtagene ciloleucel; HCRU = health care resource use; ICER = incremental cost-effectiveness ratio; LY = life-year; QALY = quality-adjusted life-year; SOC = standard of care.

Table 13: Disaggregated Summary of CADTH’s Reanalysis B

Parameter

Axi-cel

SOC

Incremental

Discounted LYs

Total

12.19

9.42

2.77

Pre-progression

7.08

1.37

5.71

Post-progression

5.11

8.05

–2.94

Discounted QALYs

Total

9.11

6.96

2.14

Pre-progression

5.41

1.09

4.31

Post-progression

3.73

5.87

–2.14

AE Disutility

–0.03

0.00

–0.03

Discounted costs ($)

Total

$622,022

$116,102

$505,920

Pre-progression

$544,274

$37,950

$506,323

Drug costs

$487,347

$28,765

$458,581

Administration costs

$41,872

$6,308

$35,564

HCRU costs

$15,055

$1,998

$13,057

AE costs

$0

$879

–$879

Post-progression

$21,513

$19,079

$2,434

Drug costs

$13,164

$6,731

$6,433

Administration costs

$1,061

$875

$186

HCRU costs

$7,287

$11,473

–$4,185

Terminal care

$56,236

$59,073

–$2,837

ICER ($/QALY)

$236,359

AE = adverse event; axi-cel = axicabtagene ciloleucel; HCRU = health care resource use; ICER = incremental cost-effectiveness ratio; LY = life-year; QALY = quality-adjusted life-year; SOC = standard of care.

Scenario Analyses

Table 14: Scenario Analysis Conducted on CADTH Reanalyses A and B

Stepped analysis

Drug

Total costs ($)

Total QALYs

ICER ($/QALY)

CADTH reanalysis A:

Exponential OS for axi-cel

SOC

$116,105

6.97

Reference

Axi-cel

$621,329

7.90

$544,875

CADTH reanalysis A, scenario 1:

Progressed disease utility

SOC

$116,105

7.03

Reference

Axi-cel

$621,329

7.92

$564,126

CADTH reanalysis B:

Log-normal OS for axi-cel

SOC

$116,105

6.97

Reference

Axi-cel

$621,991

9.05

$243,879

CADTH reanalysis B, scenario 1:

Progressed disease utility

SOC

$116,105

7.03

Reference

Axi-cel

$621,991

9.08

$246,379

Axi-cel = axicabtagene ciloleucel; ICER = incremental cost-effectiveness ratio; OS = overall survival; QALY = quality-adjusted life-year.

Appendix 5: Submitted BIA and CADTH Appraisal

Note that this appendix has not been copy-edited.

Table 15: Summary of Key Take-Aways

Key take-aways of the BIA

  • CADTH identified the following limitations in the sponsor’s base case: the projected market share of axi-cel is underestimated, the proportion of patients who receive second-line therapy is underestimated, the proportion of patients who receive active therapy in third-line therapy is underestimated, and CAR T-cell pretreatment costs are underestimated.

  • CADTH conducted reanalyses of the BIA by adjusting the projected share of axi-cel and increasing the proportion of patients with FL who would relapse and continue with treatment in second-line.

  • Based on the CADTH base case, the estimated budget impact associated with the reimbursement of axi-cel for the treatment of r/r grade 1, 2, or 3a FL after ≥ 2 lines of systemic therapy is expected to be $36,353,386 in year 1, $74,624,909 in year 2, and $99,608,235 in year 3, with a 3-year total of $210,586,531, under the drug plan perspective. When considering a health care system perspective, the CADTH base case estimated a budgetary impact of $38,924,621 in year 1, $79,905,269 in year 2, and $106,624,743 in year 3, for a 3-year cumulative total of $225,454,632.

  • Under the drug plan perspective, a scenario analysis based on the assumption that 80% of patients with r/r FL would receive active therapy in third line resulted in an increase of axi-cel’s estimated 3-year budget impact to $280,782,041. This indicates that the budget impact is highly sensitive to the estimation of the patient population that is likely to seek treatment.

  • Under a health care system perspective, a scenario analysis that applied CAR T-cell therapy eligibility assessment costs uniformly across all patients starting treatment in third line resulted in an increase of axi-cel’s estimated 3-year budget impact to $232,487,999.

Summary of Sponsor’s BIA

The sponsor submitted a BIA to estimate the incremental 3-year budget impact of reimbursing axi-cel for the treatment of adult patients with r/r grade 1, 2, or 3a FL after 2 or more lines of systemic therapy, as per its Health Canada indication. The analysis was performed from the perspective of the Canadian public drug plan formulary, with a scenario analysis based on the health care system perspective. The sponsor estimated the budget impact by comparing 2 scenarios: a reference scenario that estimated the total costs associated with SOC for the treatment of adult patients with r/r grade 1, 2, or 3a FL after 2 or more lines of systemic therapy; and a new drug scenario, where axi-cel is funded in the third-line setting. SOC was composed of 50% chemoimmunotherapy (i.e., BR, CHOP, CVP, GB, GDP, and R-CVP); 12% SCT; 5% phosphoinositide 3-kinase inhibitors (i.e., idelalisib); and 33% investigational therapies offered though clinical trials. The sponsor estimated the eligible population using an epidemiology-based approach, leveraging data from multiple sources in the scientific literature39-41 and assumptions based on clinical expert input. Under the drug plan perspective, the sponsor included drug acquisition costs, as well as costs associated with pretreatment pertaining to CAR T-cell therapy (i.e., bridging and lymphodepleting therapies). In addition, the sponsor included drug administration costs (i.e., leukapheresis, hospitalization, chair time) and resource use costs in a scenario that assessed the broader budgetary impact of funding axi-cel on the health care system. The dosing modelled for axi-cel reflected the product monograph. Key inputs to the BIA are documented in Table 15.

Table 16: Summary of Key Model Parameters

Parameter

Sponsor’s estimate

(reported as year 1 / year 2 / year 3, if appropriate)

Target population

At-risk populationa

30,410,851

Incidence of FL39-41

0.00721%

Patients with r/r FL in third lineb

30.95%

Population of interest (intention-to-treat)

100%

Patients receiving active therapyb

60%

Number of patients eligible for axi-cel

423 / 440 / 457

Market uptake (3 years)

Uptake (reference scenario)

  SOC

100% / 100% / 100%

Uptake (new drug scenario)

  Axi-cel

  SOC

| || || || || || || || || || || |

| || || || || || || || || || || |

Cost of treatment (per patient)

Axi-cel (one-time)

  Acquisition

  Leukapheresisc

  Bridging therapy (weighted)

  Conditioning chemotherapy (weighted)

$485,021

$2,688

$25

$2,300

SOC

  Chemotherapy (weighted)

  auto-SCT (weighted)

  Allo-SCT (weighted)

  Idelalisib (weighted)

  Clinical trial (weighted)

$15,650

$4,226

$5,520

$3,370

$0

allo-SCT = allogeneic stem cell transplant; auto-SCT = autologous stem cell transplant; axi-cel = axicabtagene ciloleucel; FL = follicular lymphoma; SOC = standard of care.

aThe at-risk population represents the pan-Canadian population and excludes Quebec, Northwest Territories, Yukon, and Nunavut.

bAssumption based on clinical expert opinion.

cLeukapheresis is included in a scenario analysis under the broader health care system perspective.

Key assumptions made by the sponsor include:

Summary of the Sponsor’s BIA Results

Results of the sponsor’s base-case BIA suggest that the incremental expenditures associated with the reimbursement of axi-cel for the treatment of adult patients with r/r grade 1, 2, or 3a FL after 2 or more lines of systemic therapy would be $10,507,766 in year 1, $21,577,008 in year 2, and $28,629,038 in year 3, for a 3-year cumulative total of $60,713,811, under the drug plan perspective. When considering a broader health care system perspective, the sponsor’s base case estimated a budgetary impact of $11,251,024 in year 1, $23,103,865 in year 2, and $30,645,989 in year 3, for a 3-year cumulative total of $65,000,877.

CADTH Appraisal of the Sponsor’s BIA

CADTH identified several key limitations to the sponsor’s analysis that have notable implications on the results of the BIA.

CADTH Reanalyses of the BIA

CADTH conducted reanalyses of the BIA by adjusting the projected share of axi-cel and increasing the proportion of patients with FL who would relapse and continue with treatment in second line, in line with clinical expert advice.

The results of the CADTH step-wise reanalysis are presented in summary format in Table 18 and a more detailed breakdown is presented in Table 19. Based on the CADTH base case, the estimated budget impact associated with the reimbursement of axi-cel for the treatment of r/r grade 1, 2, or 3a FL after 2 or more lines of systemic therapy is expected to be $36,353,386 in year 1, $74,624,909 in year 2, and $99,608,235 in year 3, with a 3-year total of $210,586,531.

Table 17: CADTH Revisions to the Submitted BIA

Stepped analysis

Sponsor’s value or assumption

CADTH value or assumption

Corrections to sponsor’s base case

None.

Changes to derive the CADTH base case

1. Projected market share of axi-cel

Year 1: | || |

Year 2: | || |

Year 3: | || |

Year 1: 10.3%

Year 2: 20.3%

Year 3: 26.2%

2. Proportion of patients with FL who would relapse and continue with treatment in 2L

31%

55%

CADTH base case

Reanalyses 1 + 2

2L = second line; axi-cel = axicabtagene ciloleucel; BIA = budget impact analysis; FL = follicular lymphoma.

Table 18: Summary of the CADTH Reanalyses of the BIA

Stepped analysis

Three-year total

Submitted base case

$60,713,811

CADTH reanalysis 1

$118,502,784

CADTH reanalysis 2

$107,892,072

CADTH base case

$210,586,531

BIA = budget impact analysis.

CADTH conducted the following additional scenario analyses to address remaining uncertainty, using the CADTH base case. Results are provided in Table 19.

  1. Assuming that 80% of patients with r/r FL would receive active therapy in the third-line setting.

  2. Exploring the budget impact associated with the reimbursement of axi-cel from a broader health care system perspective.

  3. Revising the cost associated with leukapheresis within a scenario analysis undertaken from a health care system perspective.

  4. Applying CAR T-cell therapy eligibility assessment costs uniformly across all patients starting treatment in third line within a scenario analysis undertaken from a health care system perspective.

  5. Assuming 31% of patients with FL would relapse and continue with 2L treatment (the sponsor’s original assumption).

Table 19: Detailed Breakdown of the CADTH Reanalyses of the BIA

Stepped analysis

Scenario

Year 0 (current situation)

Year 1

Year 2

Year 3

Three-year total

Submitted base case

Reference

$11,712,343

$12,573,023

$13,483,021

$14,008,858

$40,064,902

New drug

$11,712,343

$23,080,789

$35,060,028

$42,637,896

$100,778,713

Budget impact

$0

$10,507,766

$21,577,008

$28,629,038

$60,713,811

CADTH base case

Reference

$20,813,534

$22,343,013

$23,960,134

$24,894,579

$71,197,726

New drug

$20,813,534

$58,696,399

$98,585,043

$124,502,814

$281,784,256

Budget impact

$0

$36,353,386

$74,624,909

$99,608,235

$210,586,531

CADTH scenario analysis 1: Assuming 80% of patients receive active therapy in 3L

Reference

$27,751,379

$29,790,684

$31,946,845

$33,192,772

$94,930,301

New drug

$27,751,379

$78,261,865

$131,446,724

$166,003,752

$375,712,341

Budget impact

$0

$48,471,181

$99,499,879

$132,810,980

$280,782,041

CADTH scenario analysis 2: Health care system perspectivea

Reference

$25,012,715

$27,148,712

$29,756,795

$31,597,782

$88,503,289

New drug

$25,012,715

$66,073,333

$109,662,063

$138,222,525

$313,957,921

Budget impact

$0

$38,924,621

$79,905,269

$106,624,743

$225,454,632

CADTH scenario analysis 3: Revision of leukapheresis cost (health care perspective)

Reference

$25,012,715

$27,148,712

$29,756,795

$31,597,782

$88,503,289

New drug

$25,012,715

$66,289,618

$110,105,609

$138,812,845

$315,208,072

Budget impact

$0

$39,140,906

$80,348,814

$107,215,063

$226,704,783

CADTH scenario analysis 4: Inclusion of CAR T-cell therapy eligibility cost (health care perspective)

Reference

$25,012,715

$27,148,712

$29,756,795

$31,597,782

$88,503,289

New drug

$25,012,715

$68,328,686

$112,005,375

$140,657,227

$320,991,288

Budget impact

$0

$41,179,974

$82,248,581

$109,059,444

$232,487,999

CADTH scenario analysis 5: 31% of patients relapse

Reference

$11,712,343

$12,573,023

$13,483,021

$14,008,858

$40,064,902

New drug

$11,712,343

$33,030,064

$55,476,492

$70,061,129

$158,567,686

Budget impact

$0

$20,457,042

$41,993,472

$56,052,271

$118,502,784

CADTH scenario analysis 6: 95% price reduction

Reference

$20,813,534

$22,343,013

$23,960,134

$24,894,579

$71,197,726

New drug

$20,813,534

$22,485,789

$24,326,282

$25,670,907

$72,482,978

Budget impact

$0

$142,776

$366,148

$776,328

$1,285,253

CADTH scenario analysis 7: 82% price reduction

Reference

$20,813,534

$22,343,013

$23,960,134

$24,894,579

$71,197,726

New drug

$20,813,534

$27,198,916

$33,991,708

$38,534,743

$99,725,367

Budget impact

$0

$4,855,903

$10,031,574

$13,640,164

$28,527,641

3L = third line; BIA = budget impact analysis; CAR = chimeric antigen receptor.

aHealth care perspective includes drug administration costs in third line (e.g., hospitalization, chair time, leukapheresis), adverse event costs, and resource use costs.

Ethics Review

Abbreviations

ALL

acute lymphoblastic leukemia

CAR

chimeric antigen receptor

CRS

cytokine release syndrome

DLBCL

diffuse large B-cell lymphoma

ECOG PS

Eastern Cooperative Oncology Group performance status

FL

follicular lymphoma

ICU

intensive care unit

ICANS

immune effector cell-associated neurotoxicity syndrome

MCL

mantle cell lymphoma

MM

multiple myeloma

r/r

relapsed or refractory

SCT

stem cell transplant

SOC

standard of care

Supplementary Ethical Considerations: Axicabtagene Ciloleucel for Follicular Lymphoma

Ethical considerations relevant to all chimeric antigen receptor (CAR) T-cell therapies in the treatment of hematological cancers are described in the Summary Report: Ethical Considerations in the Use of CAR T-Cell Therapies for Hematological Cancers. Ethical considerations specific to the use of axicabtagene ciloleucel (Yescarta) for the treatment of adult patients with relapsed or refractory (r/r) grade 1, 2, or 3a follicular lymphoma (FL) after 2 or more lines of systemic therapy have also been identified from a review of patient and clinician group and drug program input, as well as consultation with clinical experts engaged by CADTH for this review and CADTH clinical and economic reviewers:

Summary Report: Ethical Considerations in the Use of CAR T-Cell Therapies for Hematological Cancers

Summary

Objectives

This report summarizes the ethical considerations common to the use of CAR T-cell therapies for the treatment of children and adults with hematological cancers in Canada, as identified in the normative and empirical literature on CAR T-cell therapies and informed by previous CADTH ethics reports of CAR T-cell therapies for hematological cancers. These reports addressed ethical considerations related to CAR T-cell therapies in the context of acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), FL, mantle cell lymphoma (MCL), and multiple myeloma (MM).1-8 Past CADTH reports drew upon published literature, consultation with clinical experts, consideration of input from patient groups, clinician groups, and drug programs, and collaboration with clinical and pharmacoeconomic review teams at CADTH. Domains of interest in this Summary Report include ethical considerations related to the therapeutic context of hematological cancers, the evidentiary basis and use of CAR T-cell therapies, and health systems. In the context of this report, any reference to CAR T-cell therapy refers to CAR T-cell therapies used to treat hematological cancers.

Key Ethical Considerations

Therapeutic Context: Hematological Cancers

Patient and caregiver experiences, as well as diagnostic and treatment pathways, vary across the different hematological cancers for which CAR T-cell therapies are available or are under development (e.g., ALL, DLBCL, FL, MCL, and MM). Nonetheless, common ethical considerations are reported across indications, including those related to the high unmet needs of the patient population and equity issues related to disparities in diagnosis, treatment, and outcomes of these cancers. Presently, CAR T-cell therapies are reimbursed, or are under consideration for reimbursement, as second-line, third-line, and fourth-line therapies for patients with r/r disease, for whom there are few or no available alternative treatments or for whom alternative treatments have failed. As a result, patients eligible for CAR T-cell therapy are usually characterized as having a high unmet need for durable, life-prolonging therapy.

Published literature, which is largely reported from the US, indicates that there are disparities in diagnosis, treatment, and outcomes across hematological cancers, especially for racialized, marginalized, and low socioeconomic groups and those residing in rural areas or far from tertiary care centres, and sometimes across age groups.1,2,5-8 Published literature concerning the distribution, incidence, treatment, and outcomes of hematological cancers in Canada is more limited, in part due to gaps in the collection of age-, sex-, and race-related demographic data in Canadian health information databases.9,10 This may limit a contextualized understanding of cancer-related disparities observed in Canada and its subnational jurisdictions.1

The clinical experts consulted during previous CADTH reimbursement reviews indicated that geography (residence in rural areas and/or far from tertiary centres) and socioeconomic status could impact the distribution of diagnosis, treatment, and outcomes for hematological cancers in Canada.1,2 They noted that disparities are more likely to be observed in access to primary care before diagnosis than once a patient is actively followed in the cancer care system. However, even in cancer care, requirements to travel and leave one’s support system and costs associated with travel, time off work, or childcare, as well as inconsistent funding and support across Canadian jurisdictions, can differentially impact patients’ and caregivers’ decision-making about treatment and care, including for CAR T-cell therapies, as will be discussed later. Disparities in outcomes between age groups have also been reported in Canada, as adults older than 70 years may have fewer therapeutic options if they are considered ineligible for common second-line or third-line treatments for hematological cancers, including allogenic SCT and autologous SCT.2

Evidence and Evaluation of CAR T-Cell Therapies

Ethical Considerations in Clinical Trial Data

During reimbursement review, CAR T-cell therapies have usually been evaluated with phase I/II or II, single-arm, open-label trials that offer only limited certainty about short-term therapeutic safety and efficacy and lack head-to-head comparative effectiveness and long-term safety, efficacy, and survival data.1-8 Uncertainty about the magnitude and duration of clinical benefit presents challenges for the assessment of clinical benefits and harms.11 Clinical experts consulted during previous CADTH reimbursement reviews of CAR T-cell therapies noted that the risks associated with evidentiary uncertainty for particular therapies are partially mitigated by the growing body of evidence on CAR T-cell therapies as a therapeutic class, which facilitates earlier identification and response to adverse events.1,2 Evidence-generating measures, such as active post-market surveillance, are required to better understand the risk-benefit profile and cost-effectiveness of CAR T-cell therapies in practice,12 and to inform the clinical and policy decision-making that serves the interests of patients and the public.11,13,14

The extent to which participants in CAR T-cell therapy trials are representative of patients in clinical practice in Canada varies. CAR T-cell therapy trials have generally tended to exclude patients with an Eastern Cooperative Oncology Group Performance Status (ECOG PS) greater than 1, which may not be reflective of clinical practice.1,2,6 Further, trials tend to include patients with a median age lower than that observed in practice, which may present challenges for the applicability of results to patients who are older and exclude patients with HIV or hepatitis B.1,6 CAR T-cell therapy trials also tend to include disproportionately higher rates of patients who are white than from other racial or ethnic groups, irrespective of disease incidence within the patient population.1,2,6 Indeed, racial and socioeconomic disparities in access to, and inclusion in, clinical trials have been reported in clinical trials for CAR T-cell therapies in the US (where most CAR T-cell trials are conducted).15,16 For example, participants who are African American or Black were underrepresented in clinical trials of 5 CAR T-cell products across 7 indications for hematological cancers, and are often underrepresented in clinical trials for cancer therapies across hematological indications more generally.1,6-8,15 This may potentially exacerbate existing health disparities observed in these populations15 and lead to a limited understanding of, and hinder efforts to eliminate, the racial and ethnic disparities observed in disease outcomes for these populations.17

The underrepresentation of racial, ethnic, and other marginalized groups, as well as women, in clinical trials has been identified as a common issue in clinical trials more generally. Underrepresentation in trial participation is ethically concerning, as diverse clinical trial participation contributes to building trust in medical research and institutions (which can impact a patient’s willingness to pursue treatment), promotes fairness for potential participants and their communities, and produces higher quality biomedical knowledge.18 Clinical experts consulted by CADTH in a previous reimbursement review were uncertain about the clinical implications of the underrepresentation of racial or ethnic groups in CAR T-cell trials.1 However, demographically representative clinical trial data for CAR T-cell therapies may help to determine whether therapeutic efficacy varies between subgroups and whether nontherapeutic factors (such as caregiver support or socioeconomic status) have an impact on effectiveness and clinical outcomes in the real world.1,19 Greater support is required to facilitate equitable access to clinical trial participation and to CAR T-cell treatment centres,15,18 and it is important to consider how trial participant selection may privilege certain groups and disadvantage others where demand for CAR T-cell therapy and trial participation exceed supply.11,20

Ethical Considerations in Economic Models

The lack of long-term safety, efficacy, and survival data, as well as head-to-head comparative effectiveness data, at the time of a reimbursement review has implications for the pharmacoeconomic assessment of CAR T-cell therapies, as it limits the ability to accurately model and assess cost-effectiveness.1,21,22 Uncertainty about pharmacoeconomic assessments, which are used to support the ethical principles of stewardship and public accountability in resource allocation,3 has implications for resource allocation at a health systems level, because it hinders assessments of opportunity costs (or forgone benefits) associated with the reimbursement and resourcing of CAR T-cell therapies over other resources.1,6,23 Data collection for long-term safety, efficacy, and comparative effectiveness may support more the robust pharmacoeconomic assessments used to inform reimbursement recommendations and decisions.23

Concerns about evidentiary limitations in pharmacoeconomic assessments and health-system sustainability have prompted consideration of alternative pricing and reimbursement models (e.g., value-based agreements, outcome-based pricing) as potential risk-sharing mechanisms that could possibly help mitigate the risks that payers face when reimbursing high-cost therapies, including CAR T-cell therapies, based on uncertain clinical and pharmacoeconomic evidence.6,23-28 Although not currently used in Canada for the reimbursement of CAR T-cell therapies, risk-sharing payment models have been used in other jurisdictions (especially in Europe).24 However, the way such financial arrangements are designed has ethical implications for the distribution of their potential benefits and burdens (e.g., for patients, the public, patients, payers, and manufacturers).28 For example, the way the value of a drug is defined, such as which surrogate outcomes are selected to evaluate efficacy, impacts how financial risks are distributed between manufacturers and payers.

The budget impact of implementing a CAR T-cell therapy may be underestimated if the estimated uptake does not reflect expected demand by patients and clinicians. In the absence of challenges related to manufacturing and delivery capacity, which will be discussed later, CAR T-cell therapies that are reimbursed are expected to be widely adopted by clinicians and patients, resulting in a high expected budget impacted.1 Higher budget impacts may present challenges for health systems with respect to the consideration of opportunity costs and fair resource allocation within and beyond the reimbursement of hematological-oncological therapies.6

Use of CAR T-Cell Therapies

Potential Benefits and Harms in the Use and Delivery of CAR T-Cell Therapies

CAR T-cell therapies have the potential to expand access to therapeutic options for patients without alternative options, including those who are ineligible for SCT (e.g., patients who are still sufficiently healthy to receive CAR T-cell therapy but not to undergo SCT, patients who could not find a suitable match for allogeneic SCT, and patients who exceed the age cut-offs for SCT). As a result, CAR T-cell therapies may offer equity-related advantages by expanding therapeutic options for older patients and for patients who are Black, Indigenous, and racialized, who may be underrepresented in SCT registries and thus unable to find adequate matches for allogeneic SCT in a timely manner.2,29 CAR T-cell therapies may offer additional practical advantages over existing therapies, especially for patients residing in rural or remote regions or with mobility issues, as they require a single infusion and treatment period, and as a durable therapy, may offer the first treatment-free window for patients with some cancers (e.g., MM).1,30,31

Nonetheless, most CAR T-cell therapies lack long-term safety and efficacy data at the time of reimbursement review, which limits the assessment of clinical benefits and harms. In practice, the balance of potential risks and benefits associated with CAR T-cell therapy is assessed relative to available alternative therapeutic options and to a patient’s condition (which, in the case of r/r cancer, may have a poor prognosis).1,11,32,33 CAR T-cell therapies bear the risk of severe toxicities, including CRS and other adverse events. Moreover, shortages or inconsistent availability of treatments (e.g., tocilizumab) used to treat patients who develop adverse events (e.g., CRS) after CAR T-cell therapy could impact the safe administration of these therapies.4

Although the long-term safety of CAR T-cell therapies remains uncertain, clinical experts consulted in previous reimbursement reviews noted that the safety of CAR T-cell therapies has improved as clinicians have become more experienced at administering treatment and identifying and responding to adverse events.1,2 This suggests that the safety of CAR T-cell therapies is context-dependent, where safety and efficacy may be impacted by the level of experience of the treating team and centre and the availability of supportive resources.12 The collection of post-market data and real-world evidence related to the use of novel CAR T-cell therapies could contribute to a more robust understanding of the real-world safety and efficacy of CAR T-cell therapies, and the balance of risks and benefits, in diverse clinical practice settings and communities.

Equitable Access to CAR T-Cell Therapies

The safe and effective administration of CAR T-cell therapies presently requires administration in a limited number of accredited treatment centres equipped with specialized infrastructure and highly trained providers, which are currently localized in large urban centres in Canada. As a result, access to CAR T-cell therapies may be moderated by geographic and financial barriers. Patients residing far from treatment centres (including in other provinces or territories) must travel to access treatment and spend more than a month near the treatment centre for pre-infusion and post-infusion treatment and care.1-3 The financial and psychosocial burdens resulting from geographic distance may impact patients’ therapeutic decision-making (e.g., patients opting for noncurative or inferior treatments to avoid leaving their communities or spending an extended time in hospital).1

Disparities in access to CAR T-cell therapies have been widely reported in the US context, including across race, geography (residence), and socioeconomic status.34,35 Geographic disparities in access to CAR T-cell therapies are especially salient in Canada, and especially for populations residing in rural and Northern communities or in provinces and territories without CAR T-cell centres, given Canada’s vast geography and the limited number of established and proposed CAR T-cell centres.1,2 In the Canadian context, race-based disparities in access should be considered, as they impact Indigenous people — especially in light of their disproportionately increased representation in rural and Northern communities — as well as other marginalized people or groups.1,2 At the same time, CAR T-cell therapies may offer access-related advantages over, and be less burdensome than, existing treatments, as they only require a single treatment period.1,31 Ensuring equitable access to high-quality care across Canada may also require considering what, if anything, might be owed to patients who are eligible for, but opt not to pursue, effective therapeutic options such as CAR T-cell therapy, due to geographic or other barriers.1

Presently in Canada, most jurisdictions provide some support for accommodation and/or food-related expenses for people who reside a certain distance from an infusion centre, whereas fewer provide support for travel costs.1 CAR T-cell manufacturers may offer programs for financial and/or accommodation support for required travel, but often include distance-related eligibility cut-offs, which could leave gaps in coverage for some patients or provide insufficient support to cover all costs borne by patients and caregivers.1,2,6,36 Adequate financial support for patients and caregivers may be important for facilitating equitable access to CAR T-cell therapies by mitigating cost-related barriers that are exacerbated by geography (e.g., costs associated with travel, accommodations, and lost income for patients and caregivers who reside outside of cities with CAR T-cell treatment facilities).1,6

Referral practices can also impact access to CAR T-cell therapies in Canada.6,12,37,38 Not only do patients require access to primary care, to be referred for CAR T-cell therapy, physicians must be aware of available therapies and eligibility criteria, as well as the processes involved in making a referral to a treatment centre (which could be located in a different jurisdiction).1,2 Providers less confident in their knowledge about CAR T-cell therapies may be less likely to refer,37 and racial and ethnic disparities observed in the distribution of patients receiving CAR T-cell therapy may be, in part, explained by disparities in referral patterns in primary care rather than in treatment practices in cancer care.38 Accordingly, it is important to have clear and equitable referral practices, educate clinicians about CAR T-cell therapies and referral processes, facilitate communication between clinicians and treatment centres, and provide systems-level supports for clinicians practising outside the large metropolitan centres where CAR T-cell centres are located.1,2 Eligibility for CAR T-cell therapy presently requires patients to have already undergone and failed several lines of therapy, but not all patients may have had access to, or been eligible for, earlier lines of therapy for reasons outside of their or their providers’ control; this may present a barrier to access to CAR T-cell therapy for a subset of patients.1,31

Cell Ownership

The collection and storage of patients’ cells during CAR T-cell manufacturing may raise questions related to patient privacy and cell ownership, particularly when manufacturers are outside of Canadian jurisdictions.1,6,39 It is important to recognize that tissue and genetic materials are valued differently by different cultural groups (e.g., Indigenous groups internationally), and that informed consent processes need to clearly detail cell processing and ownership, as well as how remaining cells that are not infused will be handled or disposed of.40 Consultation with diverse groups has been identified as essential to CAR T-cell research and implementation to ensure that cell handling and disposal practices, as well as educational and consent materials, are sensitive to the needs and values of diverse patients and communities.6,39,40 In the Canadian context, attention should be paid to understanding Indigenous communities’ values and practices with respect to cell and tissue ownership and governance (e.g., with reference to guidance such as the First Nations principles of OCAP [ownership, control, access, possession]).41

Considerations for Informed Consent

Processes should be in place to ensure that patients (and caregivers) are apprised of the unique risks of, and evidentiary uncertainties related to, CAR T-cell therapies to support robust and ongoing, iterative informed consent, including as patients transition between care settings.6,42-45 Robust consent processes should recognize both the unique vulnerabilities of patients with cancer who have limited or no alternative therapeutic options, and who may be exposed to hype or the underreporting of treatment-related harms or uncertainties related to CAR T-cell therapies, as well as their autonomous decision-making capacity.4,6,8 The term “cure” should be avoided in discussions to avoid misleading or promoting false hope for therapies for which long-term clinical effectiveness remains unknown.46 The balance of potential risks and benefits associated with CAR T-cell therapy should be assessed in a process of shared decision-making by patients, providers, and caregivers. For CAR T-cell therapies approved for use in pediatric populations, it is important to recognize the unique vulnerability of children who are reliant on parents or caregivers for decision-making, as well as broader support. Depending on age or determined level of competency, minors may have a more active role in consent or assent to treatment, supported by age-appropriate educational materials about the potential benefits and harms of CAR T-cell therapy to facilitate family-based discussions.43,45 Discussions related to the preservation of fertility may also be important for adolescents and young adults considering CAR T-cell therapy.2 Studying and considering patient reported outcomes and patient experiences may better facilitate shared decision-making about the use of CAR T-cell therapies.12 Additional resources, including the use of translators and the provision of age-appropriate and language-appropriate educational materials for patients and caregivers, may be required to support patient decision-making.45

Health Systems

Manufacturing and Health Systems Capacity

There are at least 2 challenges related to CAR T-cell therapy delivery in Canada: manufacturing and health systems capacity.12 The first concerns the capacity to manufacture and supply CAR T-cell therapies, and for timely coordination between manufacturers and CAR T-cell centres for limited manufacturing slots and a multiweek preparatory and manufacturing period (e.g., stabilizing patients’ conditions before apheresis, manufacturing and treatment, coordinating bridging therapy, apheresis, and the transport of cells). As each step in the complex sequence of manufacturing and delivery requirements for CAR T-cell therapy represents an opportunity for disruption or delay, it may be important to consider the development of contingency plans to ensure a stable supply.1,47 Patients may be harmed by delays in access to therapy, because they have to be in sufficiently stable and in good health to remain eligible for, and to be able to withstand, treatment.1,31 The proliferation of CAR T-cell therapies also presents a growing administrative burden for centres, which must maintain resource-intensive accreditations and manage multiple protocols for the preparation and delivery of a growing number of therapies.1 The possibility of domestic, local CAR T-cell manufacturer in hospital and research settings is currently under investigation in the CLIC-01 clinical trial in British Columbia.48 Although still nascent, the potential use of a local CAR T-cell manufacturer in the future may expedite access to CAR T-cell therapies for patients (including eliminating the time required to transport cells to and from international manufacturing facilities) and is expected to be less costly and more cost-effective than CAR T-cell therapies produced by pharmaceutical manufacturers.48

The second challenge concerns the health systems capacity required to meet the therapeutic demand for CAR T-cell therapies in Canada due to the complex infrastructure and personnel requirements.6,39 For example, implementation requires tertiary medical centres with specialized expertise; specialized training for staff; infrastructure modifications; close interactions among experienced inpatient, intensive care unit (ICU), outpatient, and emergency personnel and facilities; and the identification of and planning for patients before and after treatment. The implementation of an increasing number of CAR T-cell therapies for a growing number of indications may exacerbate existing health systems capacity challenges. Presently, there are a limited number of pediatric and adult CAR T-cell centres in Canada, which are localized in large urban centres in only some provinces. Although access in provinces and territories lacking CAR T-cell centres is managed through interjurisdictional agreements, the distribution of CAR T-cell centres in Canada could present a barrier for access to treatment for patients residing far from, or in jurisdictions without, CAR T-cell facilities. As a result, it is important to consider the allocation of CAR T-cell centres in a way that reflects regional, rural-urban, and sociodemographic equity.6,49

Although not currently used, outpatient delivery of CAR T-cell therapies has been suggested as a potential mechanism to address capacity limitations and expand access to a greater number of patients by circumventing limitations in inpatient capacity (e.g., health human resources, hospital beds, ICU capacity, apheresis facilities) and to reduce health systems costs.1,49 However, outpatient delivery would increase the need for patients to have access to social supports and a reliable caregiver, because the responsibility for care would be shifted largely onto patients and caregivers and away from trained health care personnel and health systems.1 Thus, a shift to outpatient delivery could potentially exacerbate burdens and the resulting inequities associated with accessing CAR T-cell therapies for patients and caregivers in lower socioeconomic strata and residing far from CAR T-cell centres, as is already observed in the context of SCTs.1 Outpatient delivery would still require significant health systems resources to deliver safe follow-up care for patients presenting with severe side effects or requiring ongoing care, emphasizing the need to invest in the infrastructure required to implement CAR T-cell therapies.6,39

Resource Allocation in the Context of Capacity Limitations

Insufficient supply or capacity to deliver CAR T-cell therapies raises ethical questions related to distributive justice (e.g., Who should be prioritized for access to a particular CAR-T-cell therapy, and why?), as well as procedural justice (e.g., Who should decide how to allocate limited resources and capacity? What constitutes a fair allocation process?).1,3,20,47,50 Fair decision-making processes and priority-setting criteria are required to inform the prioritization of patients for access to CAR T-cell therapies within and across indications to facilitate the equitable allocation of limited resources in Canada.1-8 Indeed, as multiple CAR T-cell therapies become available for single indications, criteria may also be required to determine whether to use 1 therapy over another,31 or whether patients would be eligible (and if so, under what conditions) for re-treatment with CAR T-cell therapy. The development of pan-Canadian priority-setting criteria for prioritizing access to CAR T-cell therapies and/or pan-Canadian coordination could facilitate fair resource allocation processes, accountability in decision-making, equitable pan-Canadian access to CAR T-cell therapies, reduce decision-making burden for clinicians, and reduce inefficiencies as a result of duplicated efforts.1,3,50 Consideration of manufacturing and health systems capacity implications may be required if CAR T-cell therapies demonstrate long-term curative potential, which could prompt the use of CAR T-cell therapy in earlier lines of treatment and, thus, for a greater number of patients.11

Funding, Opportunity Costs, and Data Infrastructure

The reimbursement and implementation of CAR T-cell therapies, which are highly expensive and resource intensive, raises concerns about the sustainability of the Canadian health care system1,6,12 and stewardship, or the responsible use of health resources based on available evidence.3 Reimbursing and implementing CAR T-cell therapies presents opportunity costs (or forgone benefits for other treatments or health care services) for fixed health care budgets in which not all services or therapies can be reimbursed, both within hematological and oncological therapies and in other therapeutic classes.12,14,23,42,51,52 Additionally, it presents opportunity costs for health systems resources (e.g., hospital beds, ICU capacity, access to clinical specialists) due to the resource-intensive nature of CAR T-cell therapies.1,3 As discussed previously, uncertainty in the clinical evidence and pharmacoeconomic models used to evaluate CAR T-cell therapies limits the ability to accurately assess the magnitude of benefit of CAR T-cell therapies relative to other treatments or services, and thus to inform an understanding of whether the benefits and burdens associated with funding some therapies or services but not others are distributed fairly.23 Clear and transparent decisions about the expansion of access to CAR T-cell therapies in the context of existing systems constraints, competing health care priorities, and long-term health systems sustainability are required to support fair decision-making and sustain patient and public trust.1,11,26,42 Although, as discussed previously, alternative pricing and reimbursement models may potentially help attenuate the risks faced by payers reimbursing therapies based on uncertain clinical and pharmacoeconomic evidence, it is still important to recognize that CAR T-cell therapies would still remain very expensive and resource intensive from a health systems perspective.1

From a health systems perspective, it is also important to consider the clinical and health informatics infrastructure and resources required to collect the data needed to implement novel funding models and post-market surveillance.14,39

Conclusion

CAR T-cell therapies are being introduced as second-line, third-line, and fourth-line therapies for the treatment of various hematological cancers (e.g., ALL, DLBCL, FL, MCL, MM). Published empirical and normative literature, as well as past CADTH ethics reviews of CAR T-cell therapies, were reviewed to identify the ethical considerations relevant to the use of CAR T-cell therapies for the treatment of hematological cancers. Ethical considerations in the context of hematological cancers include the need for an effective, durable treatment that prolongs life, as well as existing disparities in the incidence, diagnosis, treatment, and outcomes for racialized, marginalized, and low socioeconomic groups, although more data are required to inform a greater understanding of disparities in the Canadian context. Clinical trials assessing CAR T-cell therapies may not be fully representative of the patient population in Canada (e.g., across race, age, and functional status) and lack long-term safety and efficacy data and comparative effectiveness data. The lack of long-term and comparative clinical data limits the certainty of pharmacoeconomic assessments, which poses challenges for the assessment of opportunity costs, and may expose payers to greater financial risks. The way alternate pricing or funding arrangements are designed has implications of the distribution of the potential benefits and risks associated with the reimbursement of high-cost therapies based on uncertain clinical and pharmacoeconomic evidence. Underestimates in the demand for CAR T-cell therapy can lead to underestimates in the total budget impact of reimbursing and implementing CAR T-cell therapies.

The implementation of CAR T-cell therapies to clinical practice raises several access-related considerations, given a limited delivery capacity and resulting geographic barriers to access; notably, barriers to access may disproportionately impact racialized, marginalized, and low socioeconomic groups, as well as those lacking caregiver support. The reimbursement and implementation of an increasing number of CAR T-cell therapies raises several ethical considerations for health systems, including challenges associated with scaling CAR T-cell delivery across Canada due to the complex and resource-intensive infrastructure and personnel requirements. A possible shift to outpatient delivery in the future may expand access to CAR T-cell therapies, but may also shift responsibility for care onto patients and caregivers, and may disproportionately burden patients without robust caregiver support. The development of fair, consistent criteria to prioritize access to CAR T-cell therapy would facilitate equitable access across Canada, especially if demand exceeds manufacturing or delivery capacity (e.g., the growing number of CAR T-cell therapies and use in earlier lines of therapy if CAR T-cell therapies demonstrate curative potential may exacerbate demand). Additionally, the high cost of implementing CAR T-cell therapies presents a challenge for health care budgets and raises questions about the systems-level opportunity costs (both within and beyond the oncological space) of reimbursing CAR T-cell therapies.

The absence of long-term and comparative evidence for the safety and efficacy of CAR T-cell therapies necessitates robust post-market surveillance to better understand the risk-benefit profile, as well as cost-effectiveness, of CAR T-cell therapies in practice. Moreover, where possible, post-market surveillance and the use of real-world evidence may contribute to a better understanding of how the safety and efficacy of CAR T-cell therapies in clinical practice may be impacted by nonclinical factors, and whether this has an impact on how the benefits and burdens associated with the use of this therapy are distributed fairly across diverse demographic subgroups of patients with hematological cancers in Canada.

References

1.Drug Reimbursement Review clinical, pharmacoeconomic, and ethics reports: ciltacabtagene autoleucel (Carvykti) for the treatment of adult patients with multiple myeloma, who have received at least three prior lines of therapy, including aproteasome inhibitor, an immunomodulatory agent and ananti-CD38 antibody, and who are refractory to their last treatment. Can J Health Technol. 2023;3(8). 10.51731/cjht.2023.706. Accessed 2023 Aug 24.

2.Drug Reimbursement Review clinical, pharmacoeconomic, and ethics reports: brexucabtagene autoleucel (Tecartus) for the treatment of adult patients with relapsed or refractory B-cell precursor acute lymphoblastic leukemia (ALL). Can J Health Technol. 2023;3(6). 10.51731/cjht.2023.668. Accessed 2023 Aug 24.

3.Tisagenlecleucel for acute lymphoblastic leukemia and diffuse large b-cell lymphoma: ethics and implementation report. (CADTH optimal use report; vol.8, no.3d). Ottawa (ON): CADTH; 2019: https://www.cadth.ca/sites/default/files/pdf/car-t/op0538-tisagenlecleucel-ethics-and-implementation-jan2019.pdf. Accessed 2023 Aug 24.

4.Axicabtagene ciloleucel for large B-cell lymphoma: ethics and implementation report. (CADTH optimal use report; vol. 9, no. 1e). Ottawa (ON): CADTH; 2019: https://www.cadth.ca/sites/default/files/pdf/car-t/ct0002-ipe-report.pdf. Accessed 2023 Aug 24.

5.Drug Reimbursement Review clinical, pharmacoeconomic, and ethics reports: brexucabtagene autoleucel (Tecartus) for mantle cell lymphoma. Can J Health Technol. 2021;1(11). 10.51731/cjht.2021.187. Accessed 2023 Aug 24.

6.Drug Reimbursement Review clinical, pharmacoeconomic, and ethics reports: axicabtagene ciloleucel (Yescarta) for treatment of adult patients with relapsed or refractory large B-cell lymphoma (LBCL), who are candidates for autologous stem cell transplant (ASCT). Can J Health Technol. 2023;3(6). 10.51731/cjht.2023.667. Accessed 2023 Aug 24.

7.Drug Reimbursement Review clinical, pharmacoeconomic, ethics, and stakeholder input reports: lisocabtagene maraleucel (Breyanzi) for relapsed or refractory large B-cell lymphoma. Can J Health Technol. 2022;2(10). 10.51731/cjht.2022.461. Accessed 2023 Aug 24.

8.Drug Reimbursement Review clinical, pharmacoeconomic, and ethics reports: idecabtagene vicleucel (Abecma) for multiple myeloma. Can J Health Technol. 2022;2(2). 10.51731/cjht.2022.250. Accessed 2023 Aug 24.

9.Tsang M, Le M, Ghazawi FM, et al. Multiple myeloma epidemiology and patient geographic distribution in Canada: a population study. Cancer. 2019;125(14):2435-2444. PubMed

10.Mohyuddin GR, Sinnarajah A, Gayowsky A, Chan KKW, Seow H, Mian H. Quality of end-of-life care in multiple myeloma: a 13-year analysis of a population-based cohort in Ontario, Canada. Br J Haematol. 2022;199(5):688-695. PubMed

11.Imbach KJ, Patel A, Levine AD. Ethical considerations in the translation of CAR-T cell therapies. Cell Gene Ther Insights. 2018;4(4):295-307.

12.Gagelmann N, Sureda A, Montoto S, et al. Access to and affordability of CAR T-cell therapy in multiple myeloma: an EBMT position paper. Lancet Haematol. 2022;9(10):e786-e795. PubMed

13.Buitrago J, Adkins S, Hawkins M, Iyamu K, Oort T. Adult survivorship: considerations following CAR T-Cell therapy. Clin J Oncol Nurs. 2019;23(2):42-48. PubMed

14.Atilla E, Kilic P, Gurman G. Cellular therapies: day by day, all the way. Transfus Apher Sci. 2018;57(2):187-196. PubMed

15.Al Hadidi S, Schinke C, Thanendrarajan S, Zangari M, van Rhee F. Enrollment of Black participants in pivotal clinical trials supporting US Food and Drug Administration approval of chimeric antigen receptor-T cell therapy for hematological malignant neoplasms. JAMA Netw Open. 2022;5(4):e228161. PubMed

16.Alqazaqi R, Schinke C, Thanendrarajan S, et al. Geographic and racial disparities in access to chimeric antigen receptor-T cells and bispecific antibodies trials for multiple myeloma. JAMA Netw Open. 2022;5(8):e2228877. PubMed

17.Marinac CR, Ghobrial IM, Birmann BM, Soiffer J, Rebbeck TR. Dissecting racial disparities in multiple myeloma. Blood Cancer J.2020;10(2):19. PubMed

18.Schwartz AL, Alsan M, Morris AA, Halpern SD. Why diverse clinical trial participation matters. N Engl J Med. 2023;388(14):1252-1254. PubMed

19.Knight JM, Hackett E, Szabo A, et al. Associations between socioeconomic status and bispecific LV20.19 CAR T-cell therapy outcomes. Haematologica. 2023;108(2):588-593. PubMed

20.Jecker NS, Wightman AG, Rosenberg AR, Diekema DS. From protection to entitlement: selecting research subjects for early phase clinical trials involving breakthrough therapies. J Med Ethics. 2017;43(6):391-400. PubMed

21.CAR T-cell therapy: perceived need versus actual evidence. Lancet Oncol. 2018;19(10):1259 PubMed

22.Jonsson B, Hampson G, Michaels J, Towse A, von der Schulenburg JG, Wong O. Advanced therapy medicinal products and health technology assessment principles and practices for value-based and sustainable healthcare. Eur J Health Econ. 2019;20(3):427-438. PubMed

23.Choi G, Shin G, Bae S. Price and prejudice? The value of chimeric antigen receptor (CAR) T-cell therapy. Int J Environ Res Public Health. 2022;19(19):12366. PubMed

24.Goncalves E. Value-based pricing for advanced therapy medicinal products: emerging affordability solutions. Eur J Health Econ.2022;23(2):155-163. PubMed

25.Holmes H. Barriers to patient access to CAR-T therapy in the community setting. Journal of Clinical Pathways. 2018; https://www.texasoncology.com/who-we-are/media-center/in-the-news/news-articles/2018/june/barriers-to-patient-access-to-car-t-therapy-in-the. Accessed 2023 Aug 24.

26.de Lima Lopes G, Nahas GR. Chimeric antigen receptor T cells, a savior with a high price. Chin Clin Oncol. 2018;7(2):21. PubMed

27.Brigand T, Prokupkova A, Muller G, ECL Access to Medicines Task Force. CAR-T cell therapies: how much for survival? Brussels (BE): Association of European Cancer Leagues; 2018: https://www.cancer.eu/wp-content/uploads/2018/06/CAR-T-ECL-Article_Final_20062018.pdf. Accessed 2023 Aug 24.

28.Kannarkat JT, Good CB, Parekh N. Value-based pharmaceutical contracts: value for whom? Value Health. 2020;23(2):154-156. PubMed

29.Apostolidou E, Lachowiez C, Juneja HS, et al. Clinical outcomes of patients with newly diagnosed acute lymphoblastic leukemia in a county hospital system. Clin Lymphoma Myeloma Leuk. 2021;21(11):e895-e902. PubMed

30.Cohen AD, Hari P, Htut M, et al. Patient perceptions regarding ciltacabtagene autoleucel treatment: qualitative evidence from interviews with patients with relapsed/refractory multiple myeloma in the CARTITUDE-1 study. Clin Lymphoma Myeloma Leuk.2023;23(1):68-77. PubMed

31.Goldfinger M, Shah N, Cooper DL. Chimeric antigen receptor T-cell therapy for patients with multiple myeloma-a call for equal opportunity. JAMA Oncol. 2023;9(3):297-298. PubMed

32.Kuehn BM. The promise and challenges of CAR-T gene therapy. JAMA. 2017;318(22):2167-2169. PubMed

33.Chimeric antigen receptor T-cell therapy for B-cell cancers: effectiveness and value - final evidence report. Boston (MA): Institute for Clinical and Economic Review (ICER); 2018: https://icer.org/wp-content/uploads/2020/10/ICER_CAR_T_Final_Evidence_Report_032318.pdf. Accessed 2023 Aug 24

34.Ahmed N, Shahzad M, Shippey E, et al. Socioeconomic and racial disparity in chimeric antigen receptor T cell therapy access. Transplant Cell Ther. 2022;28(7):358-364. PubMed

35.Emole J, Lawal O, Lupak O, Dias A, Shune L, Yusuf K. Demographic differences among patients treated with chimeric antigen receptor T-cell therapy in the United States. Cancer Med. 2022;08:08.

36.Snyder S, Albertson T, Garcia J, Gitlin M, Jun MP. Travel-related economic burden of chimeric antigen receptor T cell therapy administration by site of care. Adv Ther. 2021;38(8):4541-4555. PubMed

37.Blue B. Socioeconomic and racial disparity in chimeric antigen receptor T cell (CART) therapy access. Transplant Cell Ther. 2022;28(7):345-346. PubMed

38.Molina A, Muffly L. CT-095 disparities and incomplete capture of race/ethnicity data in adults with hematologic malignancies referred for cellular therapies. Clin Lymphoma Myeloma Leuk. 2022;22(Suppl 2):S433-S434.

39.Lamprecht M, Dansereau C. CAR T-cell therapy: update on the state of the science. 2019;23(2):6-12.

40.Weinkove R, George P, Ruka M, Haira TH, Giunti G. Chimeric antigen receptor T-cells in New Zealand: challenges and opportunities. N Z Med J. 2021;134(1542):96-108. PubMed

41.First Nations Information Governance Centre (FNIGC). The First Nations Principles of OCAP®. https://fnigc.ca/ocap-training/. Accessed 2023 Aug 24.

42.Cossu G, Birchall M, Brown T, et al. Lancet Commission: stem cells and regenerative medicine. Lancet. 2018;391(10123):883-910. PubMed

43.Mahadeo KM, Khazal SJ, Abdel-Azim H, et al. Management guidelines for paediatric patients receiving chimeric antigen receptor T cell therapy. Nat Rev Clin Oncol. 2019;16(1):45-63. PubMed

44.Darrow JJ, Sarpatwari A, Avorn J, Kesselheim AS. Practical, legal, and ethical issues in expanded access to investigational drugs. N Engl J Med. 2015;372(3):279-286. PubMed

45.McConville H, Harvey M, Callahan C, Motley L, Difilippo H, White C. CAR T-cell therapy effects: review of procedures and patient education. Clin J Oncol Nurs. 2017;21(3):E79-E86.

46.Bach PB, Giralt SA, Saltz LB. FDA approval of tisagenlecleucel: promise and complexities of a $475 000 cancer drug. JAMA. 2017;318(19):1861-1862. PubMed

47.Couzin-Frankel J. For experimental cancer therapy, a struggle to ensure supply keeps up with demand. Science 2017; https://www.sciencemag.org/news/2017/06/experimental-cancer-therapy-struggle-ensure-supply-keeps-demand. Accessed 2023 Aug 24.

48.Kekre N, Hay KA, Webb JR, et al. CLIC-01: Manufacture and distribution of non-cryopreserved CAR-T cells for patients with CD19 positive hematologic malignancies. Front Immunol. 2022;13:1074740. PubMed

49.Snyder S, Chung KC, Jun MP, Gitlin M. Access to chimeric antigen receptor T cell therapy for diffuse large B cell lymphoma. Adv Ther. 2021;38(9):4659-4674. PubMed

50.Unguru Y, Fernandez CV, Bernhardt B, et al. An ethical framework for allocating scarce life-saving chemotherapy and supportive care drugs for childhood cancer. J Natl Cancer Inst. 2016;108(6):djv392. PubMed

51.Gumer JM. A new frontier in the fight against cancer (that is, for those who can afford it): the approval and cost of CAR T-cell therapies. Voices Bioeth. 2018;4. https://journals.library.columbia.edu/index.php/bioethics/article/view/6024/2987. Accessed 2023 Aug 24.

52.Gonzalez-Vicent M, Sanz J, Fuster JL, et al. Autoimmune hemolytic anemia (AIHA) following allogeneic hematopoietic stem cell transplantation (HSCT): a retrospective analysis and a proposal of treatment on behalf of the Grupo Espanol De Trasplante de Medula Osea en Ninos (GETMON) and the Grupo Espanol de Trasplante Hematopoyetico (GETH). Transfus Med Rev.2018;32(3):179-185. PubMed

Stakeholder Input

Patient Input

Lymphoma Canada

About Lymphoma Canada

Lymphoma Canada (LC) is a national Canadian registered charity whose mission it is to empower patients and the lymphoma community through education, support, advocacy, and research. Based out of Mississauga (ON), we collaborate with patients, caregivers, healthcare professionals, and other organizations and stakeholders, to promote early detection, find new and better treatments for lymphoma patients, help patients access those treatments, learn about the causes of lymphoma, and work together to find a cure.

Resources are provided in both English and French. www.lymphoma.ca

Information Gathering

The data presented in this submission was collected from an online anonymous patient survey, which Lymphoma Canada created and promoted between April 21, 2022, to April 3, 2023. This survey was originally created in 2022, but promotion was halted after being notified the submission timeline would be delayed. In 2023, the link was re-promoted via e-mail to patients registered in the LC national emailing list and made available via social media outlets, including Twitter, Instagram, and Facebook accounts. The survey had a combination of multiple-choice, rating, and open-ended questions. Skipping logic was built into the survey so that respondents were asked questions only relevant to them. Open-ended responses were noted in this report verbatim, to provide a deeper understanding of patient perspectives. 143 responses were collected, three patients reported having experience with axicabtagene ciloleucel (Yescarta).

A summary of the demographics for those that completed LC’s survey can be found in Tables 1 to 4. The majority of patients lived in Canada (86%), are between the age of 55 and 64 (71%), female (64%), and were diagnosed with follicular lymphoma 3-5 years ago (34%).

Table 1: Country of Respondents From Lymphoma Canada Survey

Respondents

CAN

USA

International

Skipped

Total

Patients with follicular lymphoma

78

7

6

52

91

Table 2: Age Range of Respondents From Lymphoma Canada Survey

Respondents

Age (years old)

Skipped

Total

35-44

45-54

55-64

65-74

75-89

Patients with follicular lymphoma

9

11

31

27

13

52

44

Table 3: Gender of Respondents From Lymphoma Canada Survey

Respondents

Gender

Female

Male

Skipped

Total

Patients with follicular lymphoma

58

33

52

91

Table 4: Number of Years Ago Respondents Were Diagnosed With Follicular Lymphoma, From Lymphoma Canada Survey

Respondents

Years

Skipped

Total

<1

1-2

3-5

5-8

9-10

Patients with follicular lymphoma

16

16

42

16

34

19

124

Disease Experience

At Diagnosis

In Lymphoma Canada’s survey, patients were asked to rate a list of physical symptoms on a scale of 1 (no impact) to 5 (significant impact) regarding their quality of life at diagnosis. The most common reported symptoms rated as a three or higher were fatigue (50%), bodily aches and pain (33%), enlarged lymph nodes (33%), indigestion (32%), and bodily swelling (21%). Common psychosocial symptoms which were present for survey respondents were anxiety/worry (84%), stress diagnosis (77%), fear of progression (70%), and difficulty sleeping (48%).

A few patient quotes are included below which capture symptoms and experiences of what it’s like getting diagnosed with follicular lymphoma:

“There is the initial shock of a serious illness and then always the fear of progression of disease and it is hard to adjust to. The watch and wait approach take time to come to terms with although good medical observation helps so much.”

“I have a 2-year-old and the fear of not being able to care for him broke my heart. Also, the fear of not seeing him grow up was the most stressful and hurtful. But I realize now that I will, and I will not let this take me down.”

“I had to quit my PhD program because the stress of the diagnosis was too much. It also triggered a flare of an autoimmune disease.”

“I did not know anything about Lymphoma and my doctor was so stressed - she was not able to give me any hope. I wish I had known more about FL and that it is possible to live well for a long time with the diagnosis.”

Current Quality of Life

Follicular lymphoma patients were also asked in this survey to rate the physical symptoms which impact their current quality of life (on a scale of 1 = no impact, to 5 = significant impact). Symptoms rated as 3 or higher included fatigue (51%), bodily aches and pains (32%), indigestion (23%), and enlarged lymph nodes (21%). Psychosocial which continue to impact follicular patients include fear of progression/relapse (67%), anxiety/worry (67%), stress of having cancer (64%), and difficulty sleeping (39%).

Daily Activities

From a list of nine factors, lymphoma patients indicated the following factors have impacted their life (at least a 3, on a scale of 1 to 5): the ability to travel (46%), ability to spend time with family & friends (41%), ability to exercise (37%), ability to concentrate (36%), and ability to work/school/volunteer (35%). When asked to include further information about these challenges, patients left the following comments:

“It takes time to adjust to having a serious illness that changes everything about your life. Having good medical care has helped me to adjust to my new normal but I am constantly aware that my life could change at any moment.”

“I have been doing follow-up appointments for the past 13 years and it seems like the health care system has trouble with chronic care. The system of follow-up appointments has been the same since the beginning, but the administrators now regularly confuse timing of scans and blood work, they didn't before.

“My life is sort of in limbo, unfortunately, knowing that my lymphoma is not curable and will come back even though I've already had 2 lines of treatment. Because I look good and take care of my health, the mental impact is hard because I don't know how long I have nor when it will strike again. It's hard to plan anything in life.”

Summary of the Disease Experience

Experiences With Currently Available Treatments

Based on survey responses, 49% of patients underwent a period of watchful waiting before starting treatment. Patients were also asked about the number of lines of therapy received to date to treat their follicular lymphoma, majority had 1 line of treatment (43%). Table 6 outlines the most common treatments received by follicular lymphoma patients in this survey. Most patients in first or second line received chemotherapy, chemoimmunotherapy, rituximab with or without bendamustine, or radiation.

Table 5: Number of Lines of Therapy Survey Respondents Received

Respondents

Have not received therapy

1

2

3 +

Skipped

Total

Patients with follicular lymphoma

20

42

19

16

46

97

21%

43%

20%

16%

100%

Table 6: Most Common Treatments Received by Respondents From Lymphoma Canada Survey

Line of Therapy

Treatment

Number of respondents

1st

Chemotherapy or Chemoimunotherapy (R-CHOP, CVP)

31

1st

Bendamustine + Rituximab

17

1st

Rituximab

8

1st

Radiation

7

2nd

Chemotherapy or Chemoimmunotherapy

10

2nd

Rituximab

9

2nd

Bendamustine + Rituximab

7

2nd

Radiation

5

In the “follicular lymphoma treatment experience” section of LC’s survey, patients were asked to rate their satisfaction with the number of treatment options available in the frontline or relapsed/refractory setting. 57% of patients indicated they were satisfied or very satisfied with treatment options available to them in the frontline setting, whereas 22% of patients were satisfied or very satisfied with their relapsed/refractory options. This suggests more treatment options are needed for those in the second or higher line of treatment.

Survey respondents were asked to rate the statement: “My current therapy (or most recent therapy) was able to manage my follicular lymphoma symptoms” on a scale of 1 (strongly disagree) to 5 (strongly agree). Most patients either strongly agreed with this statement (40%) or strongly disagreed (20%). Top factors rated by patients as having a significant negative impact (5 out of 5) included treatment-related fatigue (28%), immediate side effects of treatment (26%), and low activity level (23%). Most common side effects reported from treatment include fatigue (69%), hair loss (41%), and constipation (38%). These results highlight follicular lymphoma treatments need to be improved to manage patient symptoms.

A few patient quotes below can be used to highlight the difficult side effects patients experience and other challenges they go through during treatment:

“When experiencing heart palpitations, fear of late or long-term side effects to heart, live with worries of fear of reoccurrence. Living with a compromised immune system. Infertility.”

“Doctors need to involve patients more in their treatment discussions and plans.”

“Seems to be a lack of knowledge here in Alberta about treatment options and emerging therapies- our oncologist wasn’t even aware car- t was approved by the FDA for follicular… worries us immensely about the treatment options being presented to us.”

Summary of Currently Available Treatments

The majority of survey respondents received one line of treatment for their FL, with chemotherapy or chemoimmunotherapy as the most common treatment regimen. 40% of patients were very satisfied with their treatment options, 20% were very unsatisfied. More treatment options are needed for FL patients in the relapsed and refractory setting.

Improved Outcomes

FL patients who completed LC’s survey rated the following factors as very important (5 out of 5) - allow me to live longer (84%), longer disease remission (82%), improve quality of life and perform daily activities (69%), control disease symptoms (63%), and normalize blood counts (58%). 68% of respondents indicated they would be willing to tolerate non-serve side effects over a short-term period when considering a novel therapy, and 42% of respondents indicated it is extremely important (10 out of 10) to have a choice in deciding which drug to take based on known side effects and expected outcomes. 79% of patients felt there is a need for more therapeutic options for FL, in terms of options to choose from and drugs proven to be effective that are accessible.

Several patients left comments when asked about expectations of novel treatments to manage their lymphoma:

“With the continual investment and donations going to cancer research, I would expect treatments to be on a continuous, positive trajectory. Adding new drugs to limit or reduce symptoms and eventually cure or prolong life should and are a occurring.”

“Would hopefully like to see new therapies and treatment options that are less toxic and have little to no side effects either late or long term. Hopefully the research is on-going to find a cure.”

“Car-T should be an available first line choice if it will lengthen the time of remission for follicular lymphoma.”

“Number one expectation is that it will extend life and have minimal short term and long-term side effects. With Covid out there (forever) it is important to minimize risk of future or ongoing infections.”

“There is a lot of studies in the US with great results than we as Canadians don’t have access to. I would love to try some of those!”

“Everyone reacts differently to treatment, if there are options for those that have a bad reaction to one treatment then it would make life much easier for them. Options are good.”

“CAR-T needs to be funded.”

Summary of Currently Available Treatments

Experience With Drug Under Review

Two FL patients completed all the Yescarta treatment questions in LC’s survey. 1 patient confirmed Yescarta treatment but skipped all other treatment questions. Of the 2 responses, both patients accessed this therapy via clinical trial, one in their second line of treatment, the other as fifth line. Side effects reported include cytokine release syndrome, neutropenia, febrile neutropenia, thrombocytopenia, constipation, and swelling.

These patients rated factors such as monitoring side-effects post-inf/sion, inability to perform daily activities, and being away from family and friends as significant (4 out of 5) or very significant (5 out of 5) impact on their physical and mental health. One person was away from home for 1-3 months, the other was away from home receiving treatment for longer than 3 months. These CAR-T patients also indicated they experienced financial challenges due to absence from work, and travel or accommodation expenses during the clinical trial. Overall, both FL patients rated their experience as good and very good, and would recommend it to other patients with R/R FL.

At Lymphoma Canada, we hear from lymphoma patients consistently, that CAR T-cell therapy should be available in earlier lines of treatment. Canadian lymphoma patients should be able to receive this treatment locally and not be expected to travel far distances to receive care. Local access will significantly improve the patient experience by reducing the fear and risk of getting sick while traveling and improving quality of life by keeping patients close to their caregivers and support systems.

Companion Diagnostic Test

Not applicable.

Anything Else?

Not applicable.

Conflict of Interest Declaration — Lymphoma Canada

To maintain the objectivity and credibility of the CADTH reimbursement review process, all participants in the drug review processes must disclose any real, potential, or perceived conflicts of interest. This Patient Group Conflict of Interest Declaration is required for participation. Declarations made do not negate or preclude the use of the patient group input. CADTH may contact your group with further questions, as needed.

Did you receive help from outside your patient group to complete this submission?

No.

Did you receive help from outside your patient group to collect or analyze data used in this submission?

No.

List any companies or organizations that have provided your group with financial payment over the past 2 years AND who may have direct or indirect interest in the drug under review.

Table 7: Financial Disclosures for Lymphoma Canada

Company

$0 to 5,000

$5,001 to 10,000

$10,001 to 50,000

In Excess of $50,000

Gilead

X

Novartis

X

Bristol Myers Squibb

X

Clinician Input

Ontario Health (Cancer Care Ontario) Hematology Cancer Drug Advisory Committee

About Ontario Health (Cancer Care Ontario) Hematology Cancer Drug Advisory Committee

OH-CCO’s Cancer Drug Advisory Committees provide timely evidence-based clinical and health system guidance on drug-related issues in support of CCO’s mandate, including the Provincial Drug Reimbursement Programs (PDRP) and the Systemic Treatment Program.

Information Gathering

Information was gathered via video conferencing and email.

Current Treatments and Treatment Goals

Current standard of care involves chemotherapy, chemo-immunotherapy, autologous stem cell transplant, and in selected patients allogeneic stem cell transplant. Radiation may also be used for symptom control and in very palliative scenarios. The disease course can be quite variable with some patients having very long remissions between therapies and others behaving in a more refractory manner.

Treatment goal is mostly palliative with some curative intent with alloSCT. There are some reports of very long-term remissions following autologous stem cell transplantation. Most important goals are delaying disease progression, improve health-related quality of life, and alleviate symptoms.

Kymriah is currently under review by CADTH, for the same indication. Toxicity profiles may differ for Yescarta and Kymriah.

Treatment Gaps (Unmet Needs)

Considering the treatment goals, please describe goals (needs) that are not being met by currently available treatments.

Patients eventually becomes chemotherapy refractory and there are no treatment options afterwards. Also repeated courses of cytotoxic therapy can be associated with marrow damage (i.e., MDS) which then limits the ability to treat further and adversely affects quality of life. CART therapy would not be expected to have long-term marrow damage issues. Although data is early, we wonder whether CART therapy might be potentially curative for some patients, compared with the currently available therapies.

Place in Therapy

How would the drug under review fit into the current treatment paradigm?

3L therapy would be an appropriate time to consider CAR T-cell therapy given the benefit of available treatment is lower for these patients. It is uncertain at this time whether this CAR-T therapy may replace autologous stem cell transplant. We suspect that CART may be tried in advance of autologous stem cell transplant in those patients who have a more chemotherapy-refractory history for their follicular lymphoma.

There will be a prevalent FL population that would be eligible for this CAR-T therapy at the time of implementation.

Which patients would be best suited for treatment with the drug under review? Which patients would be least suitable for treatment with the drug under review?

Most suitable patients would be as per the clinical trial population. Exclusion may include severe organ dysfunction and poor performance status, uncontrolled infections.

Despite being excluded in the pivotal study, we would like to consider CAR-T in selected patients who had received prior CD19- directed therapy or allogeneic stem cell transplant. CART therapy might be preferred to be used prior to autologous stem cell transplantation in some patients.

There should be some flexibility around ECOG or KPS status.

There is an existing CAR-T therapy network in Ontario that can handle patient referrals.

What outcomes are used to determine whether a patient is responding to treatment in clinical practice? How often should treatment response be assessed?

As per standard lymphoma response criteria.

What factors should be considered when deciding to discontinue treatment with the drug under review?

Not applicable as this is a single infusion. Some patients may become ineligible for therapy during CAR-T cell manufacturing.

What settings are appropriate for treatment with [drug under review]? Is a specialist required to diagnose, treat, and monitor patients who might receive [drug under review]?

Centers that have expertise in CAR T-cell therapy.

Additional Information

Not applicable.

Conflict of Interest Declarations — Ontario Health (Cancer Care Ontario) Hematology Cancer Drug Advisory Committee

To maintain the objectivity and credibility of the CADTH drug review programs, all participants in the drug review processes must disclose any real, potential, or perceived conflicts of interest. This conflict of interest declaration is required for participation. Declarations made do not negate or preclude the use of the clinician group input. CADTH may contact your group with further questions, as needed. Please refer to the Procedures for CADTH Drug Reimbursement Reviews (section 6.3) for further details.

Did you receive help from outside your clinician group to complete this submission? If yes, please detail the help and who provided it.

OH-CCO provided secretariat function to the group.

Did you receive help from outside your clinician group to collect or analyze any information used in this submission?

No.

List any companies or organizations that have provided your group with financial payment over the past two years AND who may have direct or indirect interest in the drug under review. Please note that this is required for each clinician who contributed to the input.

Declaration for Clinician 1

Name: Dr. Tom Kouroukis

Position: Lead, OH-CCO Hematology Cancer Drug Advisory Committee

Date: 23-02-2023

Table 8: COI Declaration for OH-CCO Hematology Cancer Drug Advisory Committee Clinician 1

Company

$0 to $5,000

$5,001 to $10,000

$10,001 to $50,000

In excess of $50,000

No COI

Declaration for Clinician 2

Name: Dr. Pierre Villeneuve

Position: Member, OH-CCO Hematology Cancer Drug Advisory Committee

Date: 29-03-2022

Table 9: COI Declaration for OH-CCO Hematology Cancer Drug Advisory Committee Clinician 2

Company

$0 to $5,000

$5,001 to $10,000

$10,001 to $50,000

In excess of $50,000

No COI