U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Dostarlimab (Jemperli): CADTH Reimbursement Review: Therapeutic area: Endometrial cancer [Internet]. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2022 Nov.

Cover of Dostarlimab (Jemperli)

Dostarlimab (Jemperli): CADTH Reimbursement Review: Therapeutic area: Endometrial cancer [Internet].

Show details

Pharmacoeconomic Review

Executive Summary

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

Table 1. Submitted for Review.

Table 1

Submitted for Review.

Table 2. Summary of Economic Evaluation.

Table 2

Summary of Economic Evaluation.

Conclusions

The pharmacoeconomic results are informed by results from the GARNET study, a single-arm, phase I trial that aimed to assess the efficacy and safety of dostarlimab among patients with deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) recurrent or advanced endometrial cancer (EC). According to the CADTH clinical review, the lack of a comparator group precludes the ability to assess the relative therapeutic benefit or safety of dostarlimab to relevant comparators (i.e., active treatment, standard of care). According to the CADTH clinical review, it is not clear from the GARNET study whether dostarlimab may improve progression-free survival (PFS). There is indirect evidence to suggest that dostarlimab may improve PFS, compared with the standard of care (chemotherapeutic regimens); however, no strong conclusions can be drawn because of many limitations in the indirect comparisons. The relative effects of dostarlimab on patient-relevant outcomes, such as overall survival (OS) and quality of life, are uncertain.

CADTH was not able to conduct a CADTH base-case analysis because of foundational limitations with the sponsor’s model and submitted clinical evidence. Notably, the comparator population was not matched by dMMR or MSI-H status and, thus, the cost-effectiveness of dostarlimab compared with second-line treatments in the indicated population is unknown. The sponsor’s choice of a partitioned survival model (PSM) may overestimate incremental quality-adjusted life-years (QALYs), and the long-term extrapolations of OS and PFS are highly uncertain. CADTH conducted an exploratory analysis, the results of which suggest that the model is highly sensitive to several model assumptions, including long-term survival extrapolation, stopping rules, and treatment-effectiveness waning after 24 months.

The sponsor submitted a model comparing the cost-effectiveness of dostarlimab with second-line treatments (pegylated liposomal doxorubicin [PLD]; doxorubicin; paclitaxel; current mix of treatments [CMT]; carboplatin plus PLD; carboplatin plus paclitaxel; and carboplatin). According to the comparative evidence in the submitted model, carboplatin plus paclitaxel was the treatment with the smallest amount of uncertainty related to OS and PFS. Based on the sponsor’s submitted base case, the probability that dostarlimab is cost-effective was 0% at a willingness-to-pay threshold of $50,000 per QALY. The sponsor’s base-case results indicate that 89% of the incremental benefit of dostarlimab was obtained beyond the single-arm trial period, when cost-effectiveness results vary widely, depending on the chosen extrapolation function. Based on the CADTH exploratory analysis, a price reduction of 83% would be needed for dostarlimab to be considered cost-effective at a willingness-to-pay threshold of $50,000 per QALY; however, this estimate is subject to limitations in the sponsor’s submission, most crucially the inappropriateness of the estimated efficacy of comparator treatments in the dMMR or MSI-H population. An additional price reduction may be warranted.

Treatment of dMMR or MSI-H EC with dostarlimab increases costs, compared with other currently available treatments. CADTH could not estimate the amount of health improvement that may result from treatment with dostarlimab because of highly uncertain clinical efficacy data and a lack of information about the efficacy of comparator treatments. Because of methodological limitations identified in the model and clinical evidence, the cost-effectiveness of dostarlimab is unknown.

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.

Two patient groups — the Canadian Cancer Society and the Division of Gynecologic Oncology at McGill University Health Centre — provided patient input for this review. Patient input raised concerns about the inconsistent availability of treatment options in different provinces (e.g., carboplatin plus paclitaxel is the only available option in Manitoba). Patients shared their interest and desire to have access to treatment options that are available outside of Canada. Patients noted that important goals of treatment include longer remission, improved quality of life, and the ability to enjoy life and loved ones outside of clinic visits. Patients reported that they wanted new treatments, like immunotherapy, to have fewer nondebilitating side effects. Patients reported that commonly experienced side effects of existing treatments include skin issues, fatigue, bladder control, stamina, vaginal bleeding after intercourse, vaginal dryness, hair loss, pain, concentration problems (i.e., chemotherapy “fog”) and arthritis. Patient input raised concerns about the affordability of treatment, especially considering the way it can interrupt daily activities.

Two registered clinicians and 4 clinician-input groups — the Society for Gynecologic Oncology of Canada; the British Columbia Cancer Provincial Gynecology Oncology Tumour Group; clinicians from the Divisions of Gynecologic Oncology at McGill and Sunnybrook Hospitals; and Ontario Health-Cancer Care Ontario — reported that the standard of care in the first-line treatment setting for advanced or metastatic disease is platinum-based chemotherapy, typically with carboplatin and paclitaxel. Registered clinicians indicated that dostarlimab fills an unmet need for an entire biomarker-defined population with recurrent disease. The current place in therapy for dostarlimab is in the second-line treatment setting, given that pembrolizumab is currently not funded, and there are currently no available nor accessible second-line treatments for Canadian patients. Dostarlimab is unlikely to be used again in a later line of therapy, whereas endocrine therapy and radiation therapy may be used in some circumstances. Among patients who respond to treatment, dostarlimab is expected to prolong disease progression, reduce severity of symptoms, improve quality of life, compared with available standard chemotherapy or hormone-based treatments.

Feedback from the drug plans identified several items for CADTH to take into consideration during the review. First, drug plans noted that there is no current standard-of-care treatment for patients who progress on platinum-containing regimens. Drug plans noted that relevant comparators include chemotherapy (combination and monotherapy) and hormonal therapy, and that the funding of relevant comparators varies across jurisdictions. Drug plans highlighted that although pembrolizumab is approved by Health Canada for this indication, it is not publicly funded in any jurisdiction. Drug plans anticipated the potential for indication creep, given patient eligibility for dostarlimab in the following scenarios: as a first-line therapy for those who received platinum-based therapy only in early-stage disease; for patients with dMMR or MSI-H with a contraindication to or no prior exposure to platinum-containing regimens; for patients who have received more than 2 lines of therapy for advanced or recurrent disease; and for those who wish to switch to dostarlimab from current systemic therapy for recurrent dMMR or MSI-H EC (with prior platinum-based therapy). Drug plans also require clarity about whether patients with an Eastern Cooperative Oncology Group Performance Status of 2 or greater are eligible for dostarlimab. Drug plans anticipate that dostarlimab will likely be less resource-intensive than other IV chemotherapy comparators with regard to chair time, blood-work frequency, and pharmacy sterile compounding time. Importantly, drug plans indicated that mismatch repair and microsatellite instability testing in EC is not standard in all jurisdictions. Drug plans further noted that additional resources (e.g., specialists) will be required to monitor and manage potential immune-mediated adverse effects. Drug plans raised concerns about the applicability of weight-based dosing of dostarlimab in Canadian clinical practice, as it is anticipated that drug wastage is likely. However, drug wastage is not expected if a flat dose schedule is applied, as outlined in the product monograph for dostarlimab. Drug plans shared concerns about the anticipated budget impact, given that treatment duration is likely to be significantly longer with dostarlimab than with currently funded chemotherapy comparators.

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

  • The sponsor incorporated several relevant comparators, including single-agent (i.e., carboplatin, paclitaxel, PLD) and combination (carboplatin plus paclitaxel, carboplatin plus PLD) chemotherapies and a mix of treatments that comprise various chemotherapies and hormonal therapies.
  • PFS and OS were modelled for the overall population. Health-related quality of life (HRQoL) was incorporated into the model by progression status.
  • The costs of dMMR and MSI-H testing were included.

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

  • In the economic model, the sponsor incorporated a fixed-dosing approach (i.e., 500 mg every 3 weeks, 1,000 mg every 6 weeks) for dostarlimab, as in the GARNET study. The sponsor did not include an option to assess the effects of weight-based dosing for dostarlimab and, as such, CADTH was unable to explore this option further. However, drug wastage of comparator treatments was incorporated, as appropriate. The cost-effectiveness of weight-based dosing for dostarlimab remains unknown.

Economic Review

The current review is for dostarlimab (Jemperli) in adults with dMMR or MSI-H advanced or recurrent EC who have been previously treated with platinum-based chemotherapy.

Economic Evaluation

Summary of Sponsor’s Economic Evaluation

Overview

Dostarlimab is indicated as monotherapy for the treatment of adults with recurrent or advanced dMMR or MSI-H EC that has progressed on or after prior treatment with a platinum-containing regimen.

The sponsor submitted a cost-utility analysis to assess the cost-effectiveness of dostarlimab as monotherapy, compared with multiple comparator treatments, including doxorubicin monotherapy, carboplatin monotherapy; PLD, paclitaxel monotherapy, carboplatin plus paclitaxel, and carboplatin plus PLD. The sponsor further included a basket comparator representing CMT (i.e., chemotherapies, hormone therapies, and radiation therapy).1 Chemotherapies considered as part of the CMT included cisplatin plus doxorubicin, carboplatin plus gemcitabine, cisplatin, cisplatin in combination with cyclophosphamide plus doxorubicin, and gemcitabine. Hormone therapies considered part of the mix of treatments included medroxyprogesterone, megestrol, tamoxifen, anastrozole, and exemestane. The modelled population is consistent with the reimbursement request and is aligned with the GARNET trial population, a multi-centre, open-label, phase I dose-escalation study involving patients with dMMR or MSI-H recurrent or advanced EC who have progressed on or after prior treatment with a platinum-containing regimen.1

Dostarlimab is supplied in single-use vials at a submitted price of $10,270.00 per injectable 10 mL vial (50 mg/mL). The recommended dosage for dostarlimab is 500 mg every 3 weeks for doses 1 to 4, followed by 1,000 mg every 6 weeks from dose 5 until disease progression or unacceptable toxicity. The dosages of all comparator treatments, including those in the CMT, were based on Cancer Care Ontario regimen monographs for doxorubicin (60 mg/m2 on day 1 and then every 21 days), for PLD (50 mg/m2 on day 1 and then every 28 days), for carboplatin plus paclitaxel (400 mg/m2 carboplatin on day 1 and then every 28 days, and 175 mg/m2 paclitaxel on day 1 and then every 21 days), for carboplatin (400 mg/m2 on day 1 and then every 28 days), and for paclitaxel (175 mg/m2 on day 1 and then every 28 days).1 The dosage regimen for carboplatin plus PLD was 400 mg/m2 carboplatin and 50 mg/m2 PLD on day 1 and then every 28 days based on Julius et al. (2013).2 The sponsor’s calculated cost (including administration costs and wastage) of dostarlimab is $10,270 for cycles 1 to 4 and $10,270 for subsequent cycles. Using similar assumptions, the sponsor estimated per-cycle costs for each comparator to be doxorubicin = $554.50, PLD = $2,495.5, carboplatin = $738.75, paclitaxel = $4,040.00, carboplatin and paclitaxel = $4,778.75, carboplatin and PLD = $3,234.26, CMT (chemotherapies) = $2,668.67, and CMT (hormone therapies) = $14.55.1

The clinical outcomes of interest were QALYs and LYs. The economic analysis was undertaken over a lifetime horizon (40 years) from the perspective of a publicly funded health care payer. Costs and outcomes were discounted at a rate of 1.5% annually.

Model Structure

A PSM was submitted to capture the long-term costs and effects associated with the natural history of recurrent or advanced dMMR or MSI-H EC over the model time horizon. The model consisted of 3 primary health states: PFS, progressive disease, and death.1 The proportion of patients in each health state at any time over the model’s time horizon was derived from nonmutually exclusive Kaplan-Meier (KM) survival curves. The modelled time cycle was 3 weeks. OS and PFS curves were derived from the GARNET trial for dostarlimab, and were used to determine the proportion of patients in each health state (Appendix 3, Figure 1).1 Specifically, all patients entered the model in the progression-free state. The proportion of progression-free patients was derived from the area under the PFS curves, whereas the proportion of patients with progressed disease was derived from the difference in the area under the curve between the OS and PFS curves.1 Progression in the GARNET trial was defined as time from the first dose to the earliest date of assessment of disease progression or death by any cause, according to the assessment of response by a blind independent central review that used Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1).1,3 OS was defined as the time from the first dose of study treatment to the date of death by any cause. Time to treatment discontinuation (TTD) was calculated using the GARNET trial to identify the proportion of patients who were alive and who remained on treatment for the full duration of the treatment cycle after the last recorded exposure. TTD accounted for treatment discontinuation due to death or cessation of treatment.1

Model Inputs

The patient cohort comprised patients with dMMR or MSI-H advanced (stage ≥ IIIB) or recurrent ECs from cohort A1 of the GARNET study who had been previously treated with platinum-based chemotherapy.3 The modelled population was comprised mainly of adult women, with an average age of 63 years, with a mean weight of 75.8 kg, and body surface area of 1.8 m2, based on the intention-to-treat population from the GARNET trial.1

Key clinical efficacy inputs (i.e., OS and PFS) and treatment duration (i.e., TTD) for dostarlimab were based on the results of the GARNET trial (i.e., data cut-off March 11, 2020). KM estimates of PFS, OS, and TTD from the trial period (median follow-up = 11.2 months) were used to fit parametric survival curves to extrapolate the treatment effect beyond the observed trial data (median follow-up = 16.3 months) for dostarlimab at the data cut-off (i.e., date of final analysis) over the model’s time horizon (40 years).1,3 Several parametric survival functions were fitted to the PFS, OS, and TTD data to determine the best fitting distribution based on diagnostic plots, goodness-of-fit statistics, visual inspection, and clinical expectations regarding long-term progression risk and survival. The chosen parametric survival distribution of PFS and of OS for dostarlimab was the generalized gamma distribution. TTD data for dostarlimab were obtained from the GARNET trial, and the parametric survival distribution chosen to extrapolate TTD for dostarlimab over the lifetime was the 1-knot spline distribution.1

Estimates of OS for the CMT and all other individual chemotherapies were based on individual patient data from the full real-world evidence (RWE) patient cohort derived from the National Cancer Registration and Analysis Service (NCRAS) database in the UK, whereas PFS and TTD for all comparators were based on pseudo individual patient data from the same cohort digitized from the KM curves.1 Specifically, PFS was modelled on the proxy outcome of time to next treatment.1 The sponsor used 2 approaches to derive the comparative efficacy of dostarlimab against each comparator treatment, either by fitting parametric survival distributions to KM data, where possible, or by applying hazard ratios — derived from naive comparisons, matching-adjusted indirect comparisons (MAICs), or inverse probability treatment weighting (IPTW) analyses — to the estimated survival distributions of each corresponding outcome for dostarlimab (i.e., PFS, OS, and TTD).1

The parametric survival distribution chosen to extrapolate PFS for PLD was log-logistic, whereas the OS for PLD was estimated by deriving a hazard ratio from an MAIC via KM OS data in the UK RWE cohort and applying it to the parametric survival distribution for dostarlimab from the GARNET trial.1 The comparative efficacy (i.e., OS) of doxorubicin versus dostarlimab was assumed to be the same as for dostarlimab versus PLD. Parametric survival distributions were fitted to the KM curves for OS and PFS for carboplatin plus paclitaxel and for carboplatin plus PLD, respectively, with the log-logistic distribution as the chosen extrapolation. Similarly, the log-logistic distribution was the chosen parametric survival distribution fitted to the KM curves for PFS for doxorubicin, PLD, and CMT, whereas the Weibull distribution and the exponential distributions were chosen for carboplatin and for paclitaxel, respectively.1 OS rates for CMT, carboplatin, and paclitaxel were estimated by applying a hazard ratio for each treatment compared with dostarlimab to the projected OS distribution for dostarlimab, under the assumption of proportional hazards, based on the RWE cohort data.

Health state utility values were estimated with a regression model that adjusted for baseline utility and progression status using HRQoL data collected in the GARNET study with the EQ-5D-5-Levels (EQ-5D-5L) questionnaire and EQ-5D-5L values from the Canadian population. The sponsor incorporated utility values in the base case that differed by health state.1 The utility weight assigned to the PFS health state (0.784) was greater than the utility weight assigned to the progressed disease health state (0.740), and the same utility weights were applied for all treatments.1 Age- and sex-specific utility decrements were further applied to model the decline in HRQoL at each age and were based on EQ-5D utility scores for adults in the general population of England.4 To model the utility impacts of severe treatment-related adverse events (grade 3 or higher), the sponsor applied 1-off, treatment-specific disutilities, which were derived from several National Institute of Health Care Excellence gynecological cancer appraisals.5-7

The model included costs related to drug-acquisition costs, drug-administration and dispensing fees, dMMR and MSI-H EC screening costs, adverse events, subsequent therapy, surgery costs, resource use for each health state, and terminal care. Drug-acquisition costs (including subsequent therapy) for chemotherapy treatments, as well as exemestane, medroxyprogesterone, and anastrozole, were obtained from the IQVIA database, whereas those for tamoxifen and megestrol were obtained from the Ontario Drug Benefit Formulary.1,8 Drug-acquisition costs associated with CMT were weighted by the distribution of patients on these therapies in the UK RWE cohort from the NCRAS database for chemotherapy treatments and health care claims data from a Canadian study for hormone treatments within the mix.1 Drug-administration costs were estimated based on the mean time required by health care personnel (pharmacist, nurse, and physician) to administer chemotherapy per patient visit, or infusion chair time. Costs associated with screening for dMMR and MSI-H were obtained from a costing study of genomic profiling of patients with non–small cell lung cancer.9 Costs associated with adverse events were obtained from the Ontario Ministry of Health and Long-Term Care costing tool.10

Summary of Sponsor’s Economic Evaluation Results

All analyses were run probabilistically (5,000 iterations for the base-case and scenario analyses). The deterministic and probabilistic results were similar. The probabilistic findings are presented below.

Base-Case Results

The sponsor’s base-case results were calculated as pairwise comparisons and as sequential analyses. In the sponsor’s sequential analyses, the CMT comparator was excluded. The sponsor submitted pairwise comparisons between dostarlimab and each individual comparator treatment. CADTH has presented the pairwise comparison for dostarlimab versus carboplatin plus paclitaxel, as it is the treatment with the highest number of observed events (i.e., least uncertainty) and is the treatment option that is next on the cost-effectiveness frontier (i.e., smallest number of incremental QALYs). All submitted pairwise comparisons are presented in Appendix 3.

In the sponsor’s base-case results of the pairwise comparisons between dostarlimab and individual comparators, incremental costs associated with dostarlimab ranged from $804,503 (compared with carboplatin plus paclitaxel) to $829,598 (compared with doxorubicin). Incremental QALYs gained ranged from 4.79 for dostarlimab versus carboplatin to 5.93 for dostarlimab versus doxorubicin or PLD. The incremental cost-effectiveness ratio (ICER) for dostarlimab ranged from $138,486 per QALY gained versus PLD to $171,989 per QALY gained versus carboplatin. Compared with CMT, incremental QALYs was 5.12 for dostarlimab and incremental costs were $816,233, with an ICER of $159,352 per QALY gained.

Results were driven by OS projections that predicted substantial differences in total LYs and increased drug-acquisition costs associated with dostarlimab. The sponsor’s submitted ICERs ranged from $138,000 to $172,000 (details in Appendix 3). Based on the sponsor’s base case, approximately 89% of the incremental benefit for dostarlimab was derived from the extrapolated period (beyond the single-arm trial period).

Table 3. Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus Carboplatin + Paclitaxel).

Table 3

Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus Carboplatin + Paclitaxel).

Sensitivity and Scenario Analysis Results

The sponsor conducted several scenario and sensitivity analyses, which included various discount rates (0% and 3%); the adoption of alternative parametric curves for the extrapolation of OS (exponential and lognormal) and PFS (1-knot spline and 2-knot spline) for dostarlimab; the assumption that the OS efficacy of dostarlimab after treatment waning is equal to that of CMT starting 5 years after treatment initiation out to 10 years; the application of alternative data sources for OS hazard ratios of comparator therapies (i.e., Flatiron Health database for CMT1; the ZoptEC clinical trial for doxorubicin11; the McMeekin et al. [2015]12 study of paclitaxel and docetaxel; and the Mazgani et al. [2008]13 study of carboplatin plus paclitaxel); removal of the cap for the number of treatment cycles for chemotherapies; exclusion of age-related disutilities; and exclusion of IV drug administration costs.

Several scenarios resulted in notable increases in the ICER, including the adoption of alternative parametric curves for the extrapolation of OS and PFS. Notably, the sponsor’s model was highly sensitive to the use of the exponential parametric distributions for OS for dostarlimab (resulting in an ICER that ranged from $218,206 per QALY gained versus PLD to $450,209 per QALY gained versus carboplatin plus paclitaxel), and the use of the lognormal parametric distribution, which resulted in an ICER that ranged from $199,122 per QALY gained versus PLD to $279,935 per QALY gained versus carboplatin plus paclitaxel.

CADTH Appraisal of the Sponsor’s Economic Evaluation

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

  • The comparative efficacy and safety of dostarlimab to relevant comparators is highly uncertain: A key limitation of the clinical efficacy (i.e., OS and PFS) data informing dostarlimab in the economic model is that it is based on the GARNET study, a nonrandomized, open-label, single-arm study without a comparator group. The primary objective of phase I (nonrandomized) studies is to document safety outcomes rather than efficacy. In the GARNET study, there were a limited number of patients in the efficacy dataset, and the interpretation of OS with dostarlimab is limited, owing to immature OS data and the short duration of follow-up. Additionally, the results for HRQoL and symptom-severity exploratory outcomes remained inconclusive, due to a number of important limitations, as identified by the CADTH clinical review. In the absence of comparative clinical evidence from the GARNET study to inform the efficacy (i.e., OS and PFS) and safety of dostarlimab, compared with comparator treatments, in the submitted model, and considering the lack of a common comparator, the sponsor undertook 6 indirect treatment comparisons (ITCs) — 3 MAIC reports and 3 IPTW reports — to assess the efficacy of dostarlimab compared with individual monotherapy and combination chemotherapy regimens. CADTH’s review of the clinical evidence identified a number of methodological limitations to the sponsor’s MAICs and IPTW analyses, which add considerable uncertainty to the cost-effectiveness analysis. Clinical experts consulted by CADTH indicated that in the absence of robust comparative data on PFS and OS, no firm conclusions could be drawn about how dostarlimab compares with other relevant treatment options.
    • Given the absence of direct evidence and the many limitations of the sponsor’s multiple ITCs, the cost-effectiveness of dostarlimab, compared with single-agent and compared with combination chemotherapies, is highly uncertain. The comparative effectiveness of dostarlimab versus relevant comparators remains highly uncertain.
  • The patient population in the comparator group was not matched for dMMR or MSI-H status and did not reflect the indicated population: The sponsor derived data on the comparator group from patient-level health data available from NCRAS, a synthetic real-world cohort of patients with advanced or recurrent EC in the UK. Although patients in cohort A1 in the GARNET trial included those with dMMR or MSI-H EC, per the indication, information on dMMR or MSI-H status was not available in the real-world cohort database. According to the clinical experts consulted by CADTH, dMMR or MSI-H status is a prognostic factor and strongly influences clinical management. This limitation in the sponsor’s submitted evidence means that the efficacy of dostarlimab, compared with all comparator treatments, is unknown for a dMMR and MSI-H population.
    • CADTH could not address this misalignment of the patient population through reanalysis. The cost-effectiveness of dostarlimab relative to all comparator treatments is unknown.
  • Model structure suggests a post-progression survival benefit: Results from the sponsor’s model suggest that dostarlimab is associated with longer survival after disease progression. Specifically, the sponsor’s results suggest a post-progression survival benefit for patients receiving dostarlimab, relative to carboplatin plus paclitaxel (the next-nearest comparator), such that roughly 42% of the incremental survival (2.84 LYs) would occur after patients have experienced disease progression and have discontinued dostarlimab (Table 13). The CADTH clinical review team noted that there is no evidence of a clear mechanism by which dostarlimab would continue to provide clinical benefit for patients with progressive disease, given that the GARNET study was a single-arm study and, therefore, a treatment effect between groups could not be assessed. The sponsor’s use of a PSM introduces structural assumptions about the relationship between PFS and OS that likely do not accurately reflect causal relationships within the disease pathway. These assumptions may produce a post-relapse survival bias that favours dostarlimab. Because of the structural independence between OS and PFS end points assumed in a PSM, extrapolations for each end point may reflect within-trial trends in the rates of progression and death. Last, the clinical experts consulted by CADTH observed that the results for carboplatin monotherapy yielded higher survival rates in this patient population than carboplatin plus docetaxel. This finding was considered implausible. Accordingly, CADTH omitted this finding from its analysis and chose the combination therapy as the comparator closest on the frontier to dostarlimab. For this reason, CADTH presented pairwise comparisons between dostarlimab and carboplatin plus paclitaxel.
    • CADTH asked the sponsor to provide additional evidence to support the implied post-progression benefit (2.84 incremental LYs and 1.98 incremental QALYs, compared with carboplatin plus paclitaxel); however, the sponsor did not provide any clinical evidence from the GARNET study to substantiate this claim. Rather, the sponsor asserted that RWE data suggested a post-progression benefit from dostarlimab would be possible because the proportion of patients likely to receive platinum-based therapy in the third-line would be higher than the proportion likely to receive currently available treatment options in the second-line; because a higher proportion of patients receiving dostarlimab in the model could receive active therapy after discontinuation; and because OS and PFS extrapolations applied in the sponsor’s base case represented the mechanism of action of a post-progression benefit for dostarlimab over time. The CADTH clinical review of the sponsor’s submitted evidence did not find evidence to support this explanation. The clinical experts consulted by CADTH noted that the sponsor’s chosen comparators did not match expected clinical practice and, therefore, did not support this explanation of the post-progression survival difference observed in the sponsor’s model. CADTH was unable to determine the extent to which the implied post-progression benefit was due to the effect of treatment with dostarlimab, structural bias in the PSM, or limitations in the comparator efficacy evidence.
  • Long-term extrapolations of the clinical efficacy (OS and PFS) of dostarlimab are likely overestimated: The sponsor fitted several parametric survival curves to extrapolate OS and PFS for patients who received dostarlimab over the lifetime time horizon (40 years), based on the observed period of the GARNET trial (median duration of follow-up = 16.3 months). According to the clinical experts consulted by CADTH, the estimates of OS and PFS beyond the observed period used in the GARNET study were unrealistically high and did not align with the anticipated prognosis for this patient population. The CADTH clinical review did not find evidence in the GARNET study to support the sponsor’s survival extrapolations.
    The sponsor also fitted parametric survival curves for OS, PFS, and TTD for each individual comparator based on the UK RWE cohort data. The clinical experts consulted by CADTH indicated that the extrapolated OS and PFS estimates were overestimated and did not align with the natural history of Canadian patients seen in clinical practice. Based on the second-line therapies commonly used in Canadian clinical practice, the experts indicated that approximately 60% to 70% of patients would likely remain alive at 6 months, 30% at 12 months, and 10% or so at 24 months, followed by a rapid decline in OS and PFS.
    • Incremental QALYs for all comparators are likely overestimated as a result of this limitation. CADTH explored alternative assumptions in exploratory analyses (refer to Appendix 4).
  • The sponsor’s choice of comparators may not reflect current standard-of-care treatments offered in Canadian clinical practice: The sponsor excluded several chemotherapies (monotherapies and combined treatment regimens) that were identified to be relevant comparators for adults with recurrent or advanced dMMR or MSI-H EC who have progressed on or after prior treatment with a platinum-containing regimen (i.e., pembrolizumab, cisplatin, carboplatin plus docetaxel, carboplatin plus doxorubicin, or cisplatin plus doxorubicin), according to the clinical experts consulted by CADTH. Among the comparator treatments that were included, the clinical experts consulted by CADTH noted that several did not reflect Canadian clinical practice. Of note, the experts stated that hormonal therapy is rarely used in the indicated population and, importantly, there is no true standard of care for second-line therapies.
    Additionally, the frequency of use of certain treatments as part of the standard of care in the second-line setting for the comparator group did not reflect Canadian clinical practice, particularly the distribution of patients who received a platinum doublet as a second-line therapy, such as carboplatin plus paclitaxel. The clinical experts consulted by CADTH noted that patients with dMMR or MSI-H disease are typically not sensitive to platinum-containing therapies and are therefore not eligible for re-treatment with platinum-containing regimens after progression. Accordingly, the frequency of use in the sponsor’s submission was not reflective of clinical practice for the indicated population. The clinical experts consulted by CADTH suggested that paclitaxel or liposomal doxorubicin would be the most frequently used comparators.
    • CADTH explored alternative assumptions in an exploratory analysis (refer to Appendix 4).
  • Patients who received dostarlimab were assumed to remain on treatment indefinitely over the lifetime time horizon, and the duration of treatment effect was assumed to last for a patient’s lifetime, without any treatment waning: In the sponsor’s base case, the sponsor did not apply a stopping rule or any treatment discontinuation criteria to patients who received dostarlimab. As such, patients were assumed to continue receiving dostarlimab indefinitely over the lifetime time horizon (40 years). Additionally, the sponsor assumed that the duration of treatment effect for dostarlimab would last for the patient’s lifetime (i.e., no treatment waning). Each of these assumptions were noted to be unlikely by the clinical experts consulted by CADTH. First, the clinical experts consulted by CADTH noted that 50% to 60% of patients were likely to discontinue treatment after 2 years, and the majority (approximately 90%) would likely be off treatment by 5 years. Second, CADTH’s clinical experts indicated that treatment-waning effects were likely to occur approximately 2 years after treatment initiation, based on their clinical experience with patients receiving immunotherapy in Canadian clinical practice.
    • CADTH explored alternative assumptions in an exploratory analysis (refer to Appendix 4).
  • Health state utility values for the progressed disease health state lacked face validity: In the economic model, health state utility values were estimated with EQ-5D-5L data collected from patients in the GARNET study. Based on the sponsor’s methodology, utility values for patients in the progressed disease health state (0.74) were similar to those for patients in the progression-free health state (0.78). The clinical experts consulted by CADTH indicated that patient quality of life typically worsens with disease progression. As such, health state utility values for the progressed disease health state lack face validity and likely overestimate patients’ post-progression quality of life in favour of dostarlimab. The magnitude of bias in favour of dostarlimab remains unknown, resulting in additional uncertainty about the impact of health state utility values on the ICER.
    • CADTH explored alternative assumptions in an exploratory analysis (refer to Appendix 4).
Table 4. Key Assumptions of the Submitted Economic Evaluation (Not Noted As Limitations in the Submission).

Table 4

Key Assumptions of the Submitted Economic Evaluation (Not Noted As Limitations in the Submission).

CADTH Reanalyses of the Economic Evaluation

Base-Case Results

As noted above, there are key limitations associated with the model structure, available clinical data for dostarlimab, and the comparative efficacy of relevant comparators. The use of the PSM in the current review is inappropriate, given that PSMs rely on mature OS data to produce reliable cost-effectiveness estimates. CADTH notes that the sponsor’s model predicts improbable estimates of incremental QALYs gained after disease progression, which was not supported by trial data. Importantly, the comparator population was based on data from a UK RWE patient cohort that was not matched for dMMR or MSI-H status and, thus, does not reflect the indicated population. Additionally, several included comparators were not reflective of clinical practice in Canada, and other relevant comparators were excluded. Although the clinical experts consulted by CADTH indicated that paclitaxel is the most commonly used second-line treatment, CADTH did not report results of this pairwise comparison due to sparse data. As noted in the summary of the sponsor’s economic evaluation, CADTH selected carboplatin plus paclitaxel as the main comparator; it is the treatment with the highest number of observed events (i.e., least uncertainty) and is next on the cost-effectiveness frontier. As such, the comparative effectiveness and safety of dostarlimab to relevant comparators is highly uncertain. The result of these limitations is that the costs and QALYs associated with the use of dostarlimab are highly uncertain. CADTH reanalyses cannot address this uncertainty in the clinical evidence. Consequently, CADTH was unable to conduct any base-case reanalysis of the sponsor’s model.

Scenario Analysis Results

Because a CADTH base-case reanalysis was not performed, price-reduction analyses were conducted using only the sponsor’s base case. As such, deterministic price-reduction analyses are subject to the key limitations of the sponsor’s model, noted above, and are based on publicly available prices of the comparator treatments. Based on the sponsor’s submitted base case, a reduction of 74% in the price of dostarlimab would be required for dostarlimab to be cost-effective at a conventional threshold of $50,000 per QALY, compared with carboplatin plus paclitaxel (Appendix 4). It is important to note that this price-reduction estimate is based on estimates of incremental OS that are likely not representative of the true incremental benefit of dostarlimab, including single-arm trial results, unmatched comparator populations, and overestimated and highly uncertain survival estimates. The directionality of bias in the sponsor’s submission suggests that the price reduction would need to be higher than the estimated 74%. Further details of this exploratory analysis are provided in Appendix 4.

Although CADTH did not conduct any formal reanalyses of the sponsor’s model, an exploratory analysis was undertaken to explore the impact that changes to model assumptions would have on the ICER. The key limitations of the sponsor’s base-case analysis noted in the CADTH appraisal of the sponsor’s economic evaluation apply to this exploratory analysis, including the fundamental limitation that there is no direct evidence to support the comparative efficacy of dostarlimab to multiple relevant comparator treatments. As such, this exploratory analysis should not be interpretated as a CADTH base case, as there remains uncertainty regarding the true effect of dostarlimab. The key insight from this exploratory analysis is that the cost-effectiveness estimate of dostarlimab is highly influenced by assumptions related to OS. Details of this exploratory analysis are provided in Appendix 4.

Issues for Consideration

  • Pembrolizumab is indicated for this patient population; however, it is currently not funded in Canada though it has received regulatory approval. As pembrolizumab is not currently funded, its use is relatively uncommon at present, as noted by drug plans; however, its use is expected to increase quickly over the next few years among patients with private insurance. The cost-effectiveness of dostarlimab compared with pembrolizumab is unknown.
  • The current submission is for the use of dostarlimab monotherapy for the treatment of adults with recurrent or advanced dMMR or MSI-H EC that has progressed on or after prior treatment with a platinum-containing regimen. Based on clinical experts’ responses to drug plan feedback, there is the potential for indication creep among patients who received platinum-based therapy only in early-stage disease, those who received more than 2 lines of therapy for advanced or recurrent disease, and those who are currently receiving systemic therapy for recurrent dMMR or MSI-H (with prior platinum-based therapy). Off-label use of dostarlimab among patients with advanced or recurrent EC is likely to result in an increased budget impact.
  • Drug plan feedback noted that dMMR and MSI-H testing is not a standard or reflexive test across jurisdictions, The clinical experts consulted by CADTH indicated that all patients with EC of any stage should receive dMMR and MSI-H testing at the time of diagnosis, and that it is not costly and is easily performed.
  • The submitted economic model did not include an option to assess a weight-based dosing regimen for dostarlimab; rather, 2 fixed-dosing regimens (i.e., 500 mg every 3 weeks and 1,000 mg every 6 weeks) were considered on the basis of the Health Canada indication and the GARNET study protocol. The clinical experts consulted by CADTH indicated that dostarlimab may be administered as either a flat-rate dose or a weight-based dose, based on the maximum capped dose derived from several pharmacokinetic studies in support of weight-based dosing of immunotherapies. As dostarlimab is an immunotherapy with monoclonal antibodies similar to pembrolizumab, weight-based dosing for dostarlimab is likely to be similarly implemented in Canadian practice, despite the absence of weight-based dosing information in the product monograph and will likely have an impact on associated drug costs. The cost-effectiveness of dostarlimab’s weight-based dosing regimen, however, is unknown.

Overall Conclusions

The CADTH review of the clinical evidence found no direct comparative evidence between dostarlimab and any relevant treatment comparator in the treatment of adults with recurrent or advanced dMMR or MSI-H EC. There is indirect evidence to suggest that dostarlimab may improve PFS, compared with standard of care (chemotherapeutic regimens); however, no strong conclusions can be drawn because of the many limitations in the indirect comparisons. The sponsor’s submitted ITCs were based on data from patients whose dMMR or MSI-H status was unknown, meaning that they are not aligned with the indication for dostarlimab. Thus, the effectiveness of dostarlimab remains highly uncertain relative to any currently reimbursed treatment.

The sponsor submitted a model comparing the cost-effectiveness of dostarlimab with second-line treatments (PLD, doxorubicin, paclitaxel, CMT, carboplatin plus PLD, carboplatin plus paclitaxel, and carboplatin). According to the comparative evidence in the submitted model, carboplatin plus paclitaxel was the next most effective treatment on the efficiency frontier, with the least uncertainty around OS and PFS parameters. The sponsor’s estimate of the pairwise ICER for dostarlimab versus carboplatin plus paclitaxel was $164,193 per QALY, and the probability that dostarlimab is cost-effective was 0% at a willingness-to-pay threshold of $50,000 per QALY.

CADTH was not able to conduct a reanalysis because of foundational limitations with the sponsor’s model and submitted clinical evidence. Notably, the efficacy of comparator treatments in dMMR or MSI-H EC patients was unknown and, thus, the cost-effectiveness of dostarlimab compared with second-line treatments in the indicated population is unknown. The sponsor’s choice of a PSM produced QALY estimates that were likely overestimated. The model was highly sensitive to long-term extrapolation assumptions about OS and PFS, with a sizable majority of incremental QALYs observed in the extrapolated period (i.e., beyond the period of the trial). The sponsor’s model predicts a post-progression survival benefit for dostarlimab that is not supported by the trial evidence. CADTH was unable to address the critical limitation regarding the apparent post-progression survival benefit because of constraints introduced by the submitted model structure. Exploratory analysis found that the model is highly sensitive to several model assumptions, including long-term survival extrapolation, treatment discontinuation, and treatment-effectiveness waning after 24 months. Based on the CADTH exploratory analysis, a price reduction of 83% would be needed for dostarlimab to be considered cost-effective at a willingness-to-pay threshold of $50,000 per QALY; however, this estimate is subject to limitations in the sponsor’s submission, most crucially the inappropriateness of the estimated efficacy of comparator treatments in the dMMR and MSI-H population. Additional price reduction may be warranted.

Treatment of dMMR or MSI-H EC with dostarlimab increases costs compared with other currently available treatments. CADTH could not estimate the amount of health improvement that may result from treatment with dostarlimab because of highly uncertain clinical efficacy data and a lack of information about the efficacy of comparator treatments. Because of the methodological limitations identified in the model and clinical evidence, the cost-effectiveness of dostarlimab is unknown.

Abbreviations

CMT

current mix of treatments

dMMR

deficient mismatch repair

EC

endometrial cancer

EQ-5D-5L

EQ-5D-5-Level

HRQoL

health-related quality of life

ICER

incremental cost-effectiveness ratio

IPTW

inverse probability treatment weighting

KM

Kaplan-Meier

MAIC

matching-adjusted indirect comparison

MSI-H

microsatellite instability-high

NCRAS

National Cancer Registration and Analysis Service

OS

overall survival

PLD

pegylated liposomal doxorubicin

PFS

progression-free survival

PSM

partitioned survival model

QALY

quality-adjusted life-year

RWE

real-world evidence

TTD

time to treatment discontinuation

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 5. CADTH Cost Comparison Table for dMMR or MSI-H Recurrent or Advanced Endometrial Cancer That Has Progressed on or Following Prior Treatment With a Platinum-Containing Regimen.

Table 5

CADTH Cost Comparison Table for dMMR or MSI-H Recurrent or Advanced Endometrial Cancer That Has Progressed on or Following Prior Treatment With a Platinum-Containing Regimen.

Appendix 2. Submission Quality

Note that this appendix has not been copy-edited.

Table 6. Submission Quality.

Table 6

Submission Quality.

Appendix 3. Additional Information on the Submitted Economic Evaluation

Note that this appendix has not been copy-edited.

Two figures describing the observed and extrapolated progression-free survival estimates on a time scale of A) 5 years, and B) 20 years. Multiple curves are shown, with Gompertz representing the highest extrapolated survival (approximately 40% at 5 years) and Exponential representing the lowest extrapolated survival (approximately 3% at 5 years).

Figure 1

Sponsor’s Estimates of Long-Term Progression-Free Survival for Dostarlimab.

Two figures describing the observed and extrapolated overall survival estimates on a time scale of A) 5 years, and B) 20 years. Multiple curves are shown, with Gompertz representing the highest extrapolated survival (approximately 45% at 5 years) and Gamma representing the lowest extrapolated survival (approximately 22% at 5 years).

Figure 2

Sponsor’s Estimates of Long-Term Overall Survival for Dostarlimab.

Detailed Results of the Sponsor’s Base Case

While the sponsor included multiple comparator treatments to assess the cost-effectiveness of dostarlimab, only the ICER for dostarlimab versus carboplatin + paclitaxel is presented below. The carboplatin + paclitaxel subgroup had the largest number of observations in the RWE survival cohort evidence used within the model. Accordingly, there is the least amount of parametric uncertainty for this comparator. Carboplatin + paclitaxel also had the highest comparative efficacy to dostarlimab (i.e., next-highest QALYs) relative to other subgroups. The true efficacy of carboplatin + paclitaxel within the indicated population remains unknown.

Table 7. Disaggregated Summary of the Sponsor’s Economic Evaluation Results.

Table 7

Disaggregated Summary of the Sponsor’s Economic Evaluation Results.

Detailed Results of the Sponsor’s Economic Evaluation

Table 8. Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus Pairwise Doxorubicin).

Table 8

Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus Pairwise Doxorubicin).

Table 9. Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus PLD).

Table 9

Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus PLD).

Table 10. Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus CMT).

Table 10

Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus CMT).

Table 11. Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus Carboplatin).

Table 11

Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus Carboplatin).

Table 12. Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus Paclitaxel).

Table 12

Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus Paclitaxel).

Table 13. Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus Carboplatin + PLD).

Table 13

Summary of the Sponsor’s Economic Evaluation Results (Dostarlimab Versus Carboplatin + PLD).

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

Note that this appendix has not been copy-edited.

Scenario Analyses

While CADTH did not conduct any formal reanalyses of the sponsor’s model, the economic review team performed an exploratory analysis to explore the impact of several key limitations on the ICER which included applying an alternate parametric survival distribution for OS; applying an alternate parametric survival distribution for PFS; applying a treatment stopping rule at 2 years with 60% of patients stopping treatment; applying a treatment-waning effect starting at 24 months; and revising the pricing of carboplatin.

Of note, the fundamental limitations in the sponsor’s model persist within this exploratory analysis. There is no direct evidence to support the comparative efficacy of dostarlimab to multiple relevant comparator treatments. Therefore, this exploratory analysis should not be interpreted as a formal CADTH reanalysis to which credence should be given to the results; in particular, the incremental QALY benefit estimated as part of this exploratory analysis remains unlikely to be representative of the true effect of dostarlimab, such that the corresponding ICER is unlikely to be reflective of the true cost-effectiveness of dostarlimab. Instead, the key insight from this exploratory analysis is that the cost-effectiveness estimate of dostarlimab is highly influenced by the uncertainty in the OS and PFS data, and treatment-waning effects applied at 24 months.

Despite the many limitations associated with the sponsor’s submitted ITCs, and that carboplatin + paclitaxel is unlikely to be used in the second-line setting for the indicated population according to the clinical experts consulted by CADTH, carboplatin + paclitaxel was selected as the comparator of choice to explore the cost-effectiveness against dostarlimab in the CADTH exploratory analysis as the number of data points available within the RWE cohort for OS and PFS were greater than other treatments and it demonstrated better efficacy relative to other treatments. However, the cost-effectiveness of dostarlimab compared with all single-agent and combination chemotherapies, and hormone therapy, is highly uncertain. Due to several limitations of the KM data for OS and PFS informed by the available clinical evidence and feedback from the clinical experts consulted by CADTH pertaining to the clinical plausibility of the long-term extrapolations for OS and PFS in the sponsor’s base case, CADTH selected alternate survival distributions for OS and PFS based on clinical plausibility as part of the exploratory analysis. Additionally, CADTH applied a stopping rule at 2 years following treatment initiation with 60% of patients discontinuing treatment. CADTH further applied a treatment-waning effect beginning at year 2, such that patients who initially received dostarlimab would experience similar treatment effects as carboplatin + paclitaxel. When an alternative parametric survival distribution (Exponential) was chosen for the long-term extrapolation of the OS data, the predicted incremental gain in life-years was 82% lower than in the sponsor’s base case, resulting in lower incremental QALYs (0.98) and a higher estimated ICER ($460,362 per QALY). When an alternative parametric survival distribution (Lognormal) was chosen for the long-term extrapolation of the PFS data, the predicted incremental gain in life-years was 82% lower than in the sponsor’s base case, resulting in lower incremental QALYs (0.96) and a higher estimated ICER ($470,639 per QALY).

Table 14. CADTH Revisions to the Submitted Economic Evaluation.

Table 14

CADTH Revisions to the Submitted Economic Evaluation.

Table 15. Summary of the Stepped Analysis of the CADTH Exploratory Analysis Results.

Table 15

Summary of the Stepped Analysis of the CADTH Exploratory Analysis Results.

CADTH undertook several scenario analyses on the CADTH exploratory analysis to determine the impact of alternative assumptions on the exploratory analysis of cost-effectiveness for dostarlimab compared with carboplatin + paclitaxel. This included:

  1. Comparison against the most relevant comparators in Canadian clinical practice according to the clinical experts consulted by CADTH: paclitaxel.
  2. No treatment waning.
  3. Lower health state utility value (0.625) based on the Progressed Disease health state utility value for the ZoptEC trial.1

The CADTH exploratory analysis was most sensitive to the cost-effectiveness comparison against paclitaxel, the most commonly used treatment in Canadian clinical practice according to CADTH’s clinical experts, which was informed by efficacy data from the sponsor’s indirect treatment comparisons.

Table 16. Summary of CADTH Exploratory Scenario Analyses.

Table 16

Summary of CADTH Exploratory Scenario Analyses.

Price-Reduction Analysis

As no formal CADTH reanalysis was performed, price-reduction analyses were conducted using only the sponsor’s base case assumptions. This deterministic analysis was subject to the key limitations of the sponsor’s model as noted in the CADTH Appraisal of the Sponsor’s Economic Evaluation section. Based on the CADTH exploratory analysis, a reduction in the price of dostarlimab by 83% would be required for dostarlimab to be cost-effective at a willingness-to-pay threshold of $50,000 per QALY compared to carboplatin + paclitaxel. It is important to note that this price-reduction estimate is based on estimates of incremental life-years (and hence QALYs) that are highly uncertain and may not be representative of the true incremental effect of dostarlimab. Consequently, the price reduction required for dostarlimab to be cost-effective remains unknown.

Table 17. Price-Reduction Analyses of Sponsor’s Base-Case and CADTH Exploratory Analysis.

Table 17

Price-Reduction Analyses of Sponsor’s Base-Case and CADTH Exploratory Analysis.

Appendix 5. Submitted BIA and CADTH Appraisal

Table 18. Summary of Key Take-Aways.

Table 18

Summary of Key Take-Aways.

Summary of Sponsor’s BIA

The sponsor submitted a budget impact analysis (BIA) estimating the incremental budget impact of reimbursing dostarlimab for patients with mismatch repair deficient (dMMR) or microsatellite instability-high (MSI-H) recurrent or advanced endometrial cancer (EC) that has progressed on or following prior treatment with a platinum-containing regimen. The BIA was undertaken from the perspective of the Canadian public drug plans over a 3-year time horizon, and the sponsor’s pan-Canadian estimates reflect the aggregated results from provincial budgets (excluding Quebec). In the reference scenario, patients were assumed to receive doxorubicin, PLD, carboplatin, carboplatin + paclitaxel, carboplatin + PLD, paclitaxel, and hormonal therapies, in the second-line setting following failure on first-line platinum-based chemotherapy. In the new drug scenario, dostarlimab was assumed to be reimbursed and prescribed as second-line therapy.24

The sponsor estimated the eligible population using an epidemiologic approach was derived via several assumptions and inputs to first estimate the incident population (i.e., newly eligible patients advanced or recurrent EC patients who meet the eligibility criteria for dostarlimab upon its introduction) and the prevalent population (i.e., previously eligible patients who would have met eligibility criteria for treatment with dostarlimab in the years before its introduction).24 The sponsor assumed that a proportion of patients eligible for treatment at the start of each year would initiate treatment in each quarter of the year, to calculate quarterly costs per year.24 This was accomplished by further providing breakdown of the proportion of patients on dostarlimab or other comparator treatment by progression status at each quarter year using PFS and OS curves for each treatment, respectively. The estimated numbers of patients on each treatment in each quarter were then multiplied by corresponding estimates of the quarterly costs of each treatment to ascertain quarterly treatment-specific costs per year.24

In the sponsor’s base case, drug-acquisition costs, dispensing fees, mark-up costs, costs of subsequent therapy, costs of screening for dMMR/MSI-H, and health care resource utilization costs (i.e., pharmacist, nurse, physician, and infusion chair time) were captured. Drug wastage was considered in calculations, with dosing based on Cancer Care Ontario’s regimen monographs. Drug-administration costs were not included.24

Key inputs to the BIA are documented in Table 19.

Table 19. Summary of Key Model Parameters.

Table 19

Summary of Key Model Parameters.

Summary of the Sponsor’s BIA Results

The sponsor estimated that the budget impact of reimbursing dostarlimab as second-line treatment for patients with dMMR/MSI-H recurrent or advanced EC would be $17,209,586 in year 1, $24,690,413 in year 2, and $28,464,128 in year 3 for a 3-year total budget impact of $70,364,128.24

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:

  • Uncertainty in the estimated population size eligible for dostarlimab: The sponsor undertook an epidemiological approach to estimate the size of the population eligible for dostarlimab. This required deriving inputs from the published literature and applying several assumptions to derive estimates for the incident and prevalent populations in a multi-step approach. The clinical experts consulted by CADTH indicated that although the final estimated population appeared to be largely reasonable, several estimates of the target population derived from the sponsor’s assumptions and inputs may be associated with some uncertainty. First, the clinical expert consulted by CADTH noted that the sponsor’s assumed annual incidence appeared to be slightly higher than expected. CADTH’s clinical expert indicated that according to 2021 statistics reported by a Canadian Cancer report,25 there will be approximately 8,000 new cases with an age-standardized incidence rate of 37.2 per 100,000 women. Additionally, 1 of the experts noted that the sponsor’s assumed proportion of patients with Stage IV progression who would receive active therapy (70%) appeared to be reasonable while the other noted that this proportion may be lower (50%). Additionally, the clinical experts consulted by CADTH indicated that the assumed proportion of patients with dMMR/MSI-H could be as high as 30%.
    • CADTH undertook several scenario analyses to explore the uncertainty in the estimated population size. In scenario analyses, CADTH explored alternate assumptions by (a) decreasing the annual incidence of uterine cancer to 37.2%; (b) decreasing the proportion of patients with Stage IV disease progression requiring active therapy to 50%; and (c) applied the alternate assumption that 30% of patients will have a dMMR or MSH-H status.
  • Exclusion of relevant comparators: As per the Health Canada indication and the sponsor’s submitted reimbursement request, the submitted pharmacoeconomic model for dostarlimab is indicated for the treatment of monotherapy for the treatment of adult patients with dMMR/MSI-H recurrent or advanced EC that has progressed on or following prior treatment with a platinum-containing regimen. Feedback from the clinical experts consulted by CADTH for this review indicates that the sponsor excluded several chemotherapy agents from the BIA (monotherapies and combined treatment regimens) that were identified to be relevant comparators for the indicated population (i.e., pembrolizumab, cisplatin, carboplatin plus docetaxel, carboplatin plus doxorubicin, and cisplatin plus doxorubicin). Additionally, the sponsor included hormonal therapies (i.e., medroxyprogesterone, megestrol, tamoxifen, anastrozole, and exemestane), which is not a relevant comparator according to the clinical experts consulted by CADTH and is commonly offered to patients with a more limited recurrence risk (e.g., low volume disease burden with an estrogen/progesterone receptor positive). As such, the incremental budget impact of dostarlimab is associated with uncertainty.
    • CADTH was unable to address this limitation.
  • The market share distribution of comparators in the reference scenario is uncertain: In the sponsor’s submitted BIA, the baseline market share distribution for the comparator treatments in the reference scenario did not reflect clinical expectations of treatments offered in the second-line setting, as indicated by CADTH’s clinical expert and drug plan feedback. CADTH’s clinical expert noted that commonly used second-line therapies in the Canadian clinical practice include paclitaxel or liposomal doxorubicin, however, in the sponsor’s base case, carboplatin + paclitaxel was assigned the largest market share while doxorubicin and paclitaxel were assigned the smallest market share. CADTH’s clinical expert further indicated that the purpose for first-line chemotherapy (either for metastatic disease, or for recurrence management) is suggestive of the expected market share distribution of second-line chemotherapy. Specifically, the clinical experts consulted by CADTH noted that patients who progress after first-line therapy do so quickly, in which case a platinum doublet such as carboplatin + paclitaxel would not be used unless patients were platinum-sensitive. Given that the indicated population is dMMR/MSI-H, these patients have aggressive disease and are platinum-refractory rather than platinum-sensitive, thus, it is unlikely for such a high proportion of patients to be retreated with carboplatin + paclitaxel.
    • CADTH did not address this limitation. Due to the uncertainty in the baseline market share distribution in the reference scenario, CADTH revised the baseline market share distribution to reflect the clinical expert opinion in a scenario analysis.
  • The anticipated uptake of dostarlimab in the new drug scenario is uncertain: The sponsor assumed that dostarlimab would have an anticipated market uptake rate of 50% in year 1, 70% in year 2, and 70% in year 3 while displacing comparator therapies proportionally to their estimated market shares in the reference scenario (i.e., market share for all other comparators would be halved in year 1, further reduced by 20% in year 2, and remain steady after that). The clinical expert consulted by CADTH indicated that while the sponsor’s anticipated uptake rate of dostarlimab in the new drug scenario appeared to be reasonable, there remains uncertainty to how dostarlimab will displace other comparators, as dostarlimab is expected to likely displace hormonal therapies and doxorubicin more than others given the low response rates of other treatments. Additionally, CADTH’s clinical expert noted that the introduction of dostarlimab would fully displace carboplatin + paclitaxel and
    • CADTH addressed this limitation by revising the market share distribution in the new drug scenario to reflect the expert’s feedback.
  • The sponsor assumed continued treatment over the 3-year time horizon, which likely overestimated drug costs: In the base case, the sponsor assumed that all patients would continue to remain on dostarlimab over the 3-year time horizon. The clinical experts consulted by CADTH indicated that approximately 50 to 60% of patients would likely stop treatment after 2 years of treatment. Based on expert feedback, CADTH revised the treatment stopping rule to 24 months, and revised the proportion of patients expected to stop treatment at 2 years to 60%.
    • CADTH addressed this limitation by permitting 60% of patients to stop treatment at 2 years.
  • The sponsor’s submitted BIA is based on the same clinical evidence (i.e., OS and PFS) as its pharmacoeconomic analysis, and may overestimate drug costs: The sponsor incorporated extrapolated OS and PFS curves up to 48 months (reflecting baseline and years 1, 2 and 3) to estimate the proportion of patients who remained alive, and those who were on treatment, respectively, in each year over the 3-year time horizon in their submitted BIA. The sponsor applied the same treatment-specific OS and PFS curves as in their pharmacoeconomic base case to calculate quarterly drug costs over the 3-year time horizon in their submitted BIA. As several limitations were identified with the efficacy data in the GARNET study, the long-term extrapolations of this clinical evidence are highly uncertain. As such, CADTH opted to use trial evidence only (i.e., Kaplan-Meier OS and PFS data from the GARNET study), as it represents OS and PFS estimates without any statistical adjustments. CADTH selected the relevant time points from this data to align with the forecasted period in the BIA. OS and PFS estimates from year 2 or 24 months onwards were assumed to remain the same for the remainder of the time horizon in the BIA.
    • CADTH addressed this limitation by applying the unadjusted Kaplan-Meier data from the GARNET study for OS and PFS in the BIA case. In a scenario analysis, CADTH explored the impact of arbitrarily assuming less efficacious estimates for OS by 20% at each time point.

CADTH Reanalyses of the BIA

Table 20. CADTH Revisions to the Submitted Budget Impact Analysis.

Table 20

CADTH Revisions to the Submitted Budget Impact Analysis.

Applying the changes in Table 20 resulted in a minor increase in the estimated budget impact under the drug plan perspective to $ over 3 years. The results of the CADTH stepwise reanalyses are presented in summary format in Table 21 and a more detailed breakdown is presented in Table 22.

Table 21. Summary of the CADTH Reanalyses of the BIA.

Table 21

Summary of the CADTH Reanalyses of the BIA.

Table 22. Detailed Breakdown of the CADTH Reanalyses of the BIA.

Table 22

Detailed Breakdown of the CADTH Reanalyses of the BIA.

CADTH conducted the following additional scenario analyses from the drug plan perspective (Scenarios 1 to 5, Table 23):

  1. Decreasing the annual incidence of uterine cancer to 37.2% based on 2021 Canadian Cancer statistics.25
  2. Applying the alternate assumption that approximately 50% of patients will have Stage IV disease with progression and require active therapy.
  3. Applied the alternate assumption that 30% of patients will have a dMMR or MSH-H status.
  4. Applied the arbitrary assumption that the overall survival is 20% less efficacious at each time point on the Kaplan-Meier OS curve.
  5. Applied an 83% reduction in the price of dostarlimab to align with the point at which the ICER is within the willingness-to-pay threshold of $50,000 per QALY in the CADTH exploratory reanalysis.

The model results were most sensitive to changes in the proportion of patients with a dMMR/MSI-H status.

Table 23. CADTH Scenario Analyses.

Table 23

CADTH Scenario Analyses.

References

1.
Pharmacoeconomic evaluation [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: Dostarlimab, 500 mg/10 mL vial (50 mg/mL) intravenous infusion. Mississauga (ON): GlaxoKlineSmith; 2021 Sept.
2.
Julius JM, Tanyi JL, Nogueras-Gonzalez GM, et al. Evaluation of pegylated liposomal doxorubicin dose on the adverse drug event profile and outcomes in treatment of recurrent endometrial cancer. Int J Gynecol Cancer. 2013;23(2):348-354. [PMC free article: PMC3899886] [PubMed: 23295941]
3.
Oaknin A, Tinker AV, Gilbert L, et al. Clinical Activity and Safety of the Anti-Programmed Death 1 Monoclonal Antibody Dostarlimab for Patients With Recurrent or Advanced Mismatch Repair-Deficient Endometrial Cancer: A Nonrandomized Phase 1 Clinical Trial. JAMA Oncol. 2020;6(11):1766-1772. [PMC free article: PMC7530821] [PubMed: 33001143]
4.
Ara R, Brazier JE. Using health state utility values from the general population to approximate baselines in decision analytic models when condition-specific data are not available. Value Health. 2011;14(4):539-545. [PubMed: 21669378]
5.
National Institute for Health and Care Excellence. Niraparib for maintenance treatment of relapsed, platinum-sensitive ovarian, fallopian tube and peritoneal cancer. (Technology appraisal guidance TA528) 2018: https://www​.nice.org.uk/guidance/ta528. Accessed 2022 Jan 6.
6.
National Institute for Health and Care Excellence. Olaparib for maintenance treatment of BRCA mutation-positive advanced ovarian, fallopian tube or peritoneal cancer after response to first-line platinum-based chemotherapy. (Technology appraisal guidance TA598) 2019: https://www​.nice.org.uk/guidance/ta598. Accessed 2022 Jan 6.
7.
National Institute for Health and Care Excellence. Rucaparib for maintenance treatment of relapsed platinum-sensitive ovarian, fallopian tube or peritoneal cancer. (Technology appraisal guidance TA611) 2019: https://www​.nice.org.uk/guidance/ta611. Accessed 2022 Jan 6.
8.
Ontario Ministry of Health, Ontario Ministry of Long-Term Care. Ontario drug benefit formulary/comparative drug index. 2021; https://www​.formulary​.health.gov.on.ca/formulary/. Accessed 2021 Dec 23.
9.
Johnston KM, Sheffield BS, Yip S, Lakzadeh P, Qian C, Nam J. Costs of in-house genomic profiling and implications for economic evaluation: a case example of non-small cell lung cancer (NSCLC). J Med Econ. 2020;23(10):1123-1129. [PubMed: 32597288]
10.
Ontario Case Costing Initiative (OCCI). Toronto (ON): Ontario Health and Long-Term Care; 2017: https://data​.ontario​.ca/dataset/ontario-case-costing-initiative-occi. Accessed 2021 Dec 23.
11.
Miller DS, Scambia G, Bondarenko I, et al. ZoptEC: Phase III randomized controlled study comparing zoptarelin with doxorubicin as second line therapy for locally advanced, recurrent, or metastatic endometrial cancer (NCT01767155). J Clin Oncol. 2018;36(15_suppl):5503-5503.
12.
McMeekin S, Dizon D, Barter J, et al. Phase III randomized trial of second-line ixabepilone versus paclitaxel or doxorubicin in women with advanced endometrial cancer. Gynecol Oncol. 2015;138(1):18-23. [PubMed: 25925990]
13.
Mazgani M, Le N, Hoskins PJ. Reuse of carboplatin and paclitaxel in patients with relapsed endometrial cancer--the British Columbia Cancer Agency experience. Gynecol Oncol. 2008;111(3):474-477. [PubMed: 18929402]
14.
Life tables, Canada, provinces and territories 1980/1982 to 2017/2019. Ottawa (ON): Statistics Canada; 2021: https://www150​.statcan​.gc.ca/n1/pub/84-537-x​/84-537-x2020001-eng.htm. Accessed 2022 Jan 6.
15.
Cancer Care Ontario. CRBP regimen: CARBOplatin. [no date][no date]: . Accessed 2021 Dec 23.
16.
CADTH Reimbursement Review: pembrolizumab (Keytruda). Can J Health Technol 2021;1(9). https://www​.cadth.ca​/sites/default/files​/DRR/2021/PC0235-Keytruda-Combined.pdf. Accessed 2022 Jan 6.
17.
Cancer Care Ontario. CISplatin. [no date][no date]: . Accessed 2021 Dec 23.
18.
Cancer Care Ontario. PACLitaxel. [no date][no date]: . Accessed 2021 Dec 23.
19.
Cancer Care Ontario. DOXOrubicin. [no date][no date]: . Accessed 2021 Dec 23.
20.
Cancer Care Ontario. CRBPPACL regimen: PACLitaxel-CARBOplatin. [no date][no date]: . Accessed 2021 Dec 23.
21.
Cancer Care Ontario. CRBPDOCE regimen: DOCEtaxel-CARBOplatin. [no date][no date]: . Accessed 2021 Dec 23.
22.
Cancer Care Ontario. CISPDOXO regimen: CISplatin-DOXOrubicin. [no date][no date]: . Accessed 2021 Dec 23.
23.
DeltaPA. Ottawa (ON): IQVIA; 2021: https://www​.iqvia.com/. Accessed 2021 Dec 23.
24.
Budget Impact Analysis [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: Dostarlimab, 500 mg/10 mL vial (50 mg/mL) intravenous infusion. Mississauga (ON): GlaxoKlineSmith; 2021 Sept.
25.
Canadian Cancer Statistics Advisory Committee. Canadian Cancer Statistics 2021. Toronto (ON): Canadian Cancer Society, Statistics Canada, Public Health Agency of Canada; 2021: https://cdn​.cancer.ca​/-/media/files/research​/cancer-statistics​/2021-statistics/2021-pdf-​.pdf?rev=2b9d2be7a2d34c1dab6a01c6b0a6a32d&hash​=01DE85401DBF0217F8B64F2B7DF43986REF. Accessed 2022 Jan 6.
Copyright © 2022 Canadian Agency for Drugs and Technologies in Health.

Except where otherwise noted, this work is distributed under the terms of a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International licence (CC BY-NC-ND), a copy of which is available at http://creativecommons.org/licenses/by-nc-nd/4.0/

Bookshelf ID: NBK602688

Views

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...