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

Clinical Review

Executive Summary

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

Table 1. Submitted for Review.

Table 1

Submitted for Review.

Introduction

Endometrial cancer (EC) is the most common gynecological cancer.1 An estimated 7,000 to 8,000 new cases of EC are diagnosed in Canada every year, with approximately 1,400 deaths annually.2,3 Approximately 80% of ECs are diagnosed at an early stage and are curable with surgery.1 Recurrence occurs in 13% to 20% of patients.4,5 The 5-year survival rate of patients diagnosed with metastatic or advanced disease is less than 20%.6 Patients with advanced, metastatic, or recurrent EC have limited effective therapeutic options after front-line standard treatment with a platinum-containing chemotherapeutic.

Mismatch repair-deficient (dMMR) and microsatellite instability-high (MSI-H) tumour status is a predictive biomarker of clinical benefit from checkpoint inhibitors, and represent approximately 25% of primary ECs and 13% to 30% of recurrent ECs.7-9 At first recurrence or primary advanced disease, response rates with platinum-based combination regimens in the first-line setting ranges from 40% to 62%.10-13 However, for patients with advanced or recurrent EC who have progressed on or after platinum-based chemotherapy, there is currently no standard second-line therapy. Single-drug chemotherapies or hormonal therapy may be administered, but these have low response rates and no clear survival benefit.14

Dostarlimab is an anti-programmed cell death protein-1 (PD-1) monoclonal antibody. It targets the cellular pathway between the PD-1 receptor and 2 ligands, PD-L1 and PD-L2, found on immune cells. Dostarlimab binds to the PD-1 receptor and blocks its interaction with PD-L1 and PD-L2, which allows the PD-1 pathway-mediated immune response and antitumour immune response to occur.15 On December 23, 2021, dostarlimab was approved by Health Canada for the treatment of adults with dMMR or MSI-H recurrent or advanced EC that has progressed on or after prior treatment with a platinum-containing regimen. The sponsor’s requested reimbursement criteria for dostarlimab align with the Health Canada–approved indication. Dostarlimab underwent review by Health Canada, which used an expedited review process (advance consideration under Notice of Compliance with conditions [NOC/c]). Dostarlimab has no other Health Canada–approved indication and has not previously been reviewed by CADTH. Dostarlimab was approved by the FDA and European Union, and is currently under review by the National Institute of Health Care Excellence (NICE) and the Scottish Medicines Consortium.16,17

Dostarlimab is available as a 500 mg IV infusion and is administered as an IV infusion over 30 minutes. The recommended dosage is 500 mg every 3 weeks for dose 1 through 4, and 1,000 mg every 6 weeks for dose 5 onward. Treatment may continue until disease progression or unacceptable toxicity. Dose reductions are not recommended, but dosing delays and discontinuation may be required based on safety and tolerability. Patients should be selected for treatment based on dMMR or MSI-H tumour status, determined by an accredited laboratory using validated testing methods.

The objective was to perform a systematic review of the beneficial and harmful effects of dostarlimab for the treatment of adults with dMMR or MSI-H recurrent or advanced EC that has progressed on or after prior treatment with a platinum-containing regimen.

Stakeholder Perspectives

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

Patient Input

Patient and caregiver input used for this review was collected by the Canadian Cancer Society. The input was based on an online survey and patient and caregiver testimonials. A total of 6 testimonials and 22 survey responses were received (20 patients with current or previous EC and 2 caregivers).

Respondents indicated that EC symptoms affected their daily activities, causing detrimental effects on their health-related quality of life (HRQoL). Respondents reported that the most significant side effects related to their current cancer treatment were issues with libido, sexual function, and fatigue. Loss of income due to absence from work and travel costs for cancer treatment were important financial barriers.

Respondents reported that they expect the following key outcomes for any treatment: better HRQoL, longer periods of remission, better affordability, better access, and fewer side effects. Eight of 22 respondents indicated that they had received direct experience with dostarlimab, either receiving it or assisting a patient who received it. All respondents indicated that dostarlimab was easier to use than other therapies because it had few to no side effects, longer intervals between doses, and a shorter infusion time.

Clinician Input

Input From Clinical Experts Consulted by CADTH

All CADTH review teams include at least 1 clinical specialist with expertise in 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). The following input was provided by 2 clinical specialists with expertise in the diagnosis and management of EC.

The clinical experts consulted by CADTH indicated that currently there are no standard, funded second-line treatment options for advanced or recurrent EC. The clinical experts agreed that patients who would most benefit from dostarlimab include those with identified dMMR or MSI-H recurrent or advanced EC. One of the clinical experts noted that dostarlimab could be used as monotherapy in first-line settings or later in the absence of effective treatments. The clinical experts noted that treatment with dostarlimab would not be suitable in patients with the following characteristics: very poor performance status; history of severe autoimmune disease; prior immunotherapy use; known uncontrolled central nervous system metastases and/or carcinomatous meningitis; poor medical risk due to a serious uncontrolled medical disorder; nonmalignant systemic disease or active infection requiring systemic therapy; or microsatellite stable (MSS) EC.

In the opinion of the clinical experts consulted by CADTH, treatment with dostarlimab should be discontinued in the case of disease progression, severe toxicity, or intolerability. The clinical experts indicated that the following outcomes would best assess response to treatment: overall survival (OS); response rate based on clinical and radiological investigation; progression-free survival (PFS); reduction of cancer burden and symptom improvement in activities of daily living; HRQoL; durability of response; and response to subsequent therapies.

In terms of clinically meaningful responses, the clinical experts recommended that in addition to clinical assessment of disease symptoms and duration of disease control, the use of standard immune-related Response Evaluation Criteria in Solid Tumours (irRECIST) for the assessment of response to immunotherapeutic treatments may be useful.

Clinician Group Input

A total of 7 clinician group inputs were submitted from the following groups: British Columbia Cancer Provincial Gynecological Oncology Tumour Group; McGill University Health Centre (MUHC), Division of Gynecologic Oncology; Ontario Health-Cancer Care Ontario (OH-CCO) Gynecological Drug Advisory Committee; Princess Margaret Cancer Centre (PMCC), Gynecologic Cancers Disease Site Group, Medical Oncology Group; Saskatchewan Cancer Agency (SCA); the Society of Gynecologic Oncology of Canada (GOC); and Sunnybrook Health Sciences Centre (SBHSC).

The views of the clinician groups were overall consistent with those of the clinical experts consulted by CADTH. The clinician groups indicated that the most important treatment goals are achieving disease control, delaying worsening of symptoms, prolonging survival, maintaining HRQoL, delaying disease progression, and an acceptable safety profile. All the clinician groups indicated that all patients with recurrent EC would benefit from an effective immunotherapy, but patients with dMMR or MSI-H subtypes would be the most likely to benefit from immune checkpoint inhibitor therapy. All groups recommended that patients diagnosed with metastatic EC should be offered platinum-based chemotherapy as first-line therapy. However, the British Columbia Cancer Provincial Gynecological Oncology Tumour Group did acknowledge that treatment with an immune checkpoint inhibitor may be an appropriate first-line therapy when chemotherapy is contraindicated or not desirable.

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 considerations for the initiation of therapy, continuation or renewal of therapy, and generalizability. The clinical experts consulted by CADTH weighed evidence from the GARNET trial (Study 4010-01-001)18 and other clinical considerations to provide responses to implementation questions posed by drug programs. Refer to Table 4 for more details.

Clinical Evidence

Pivotal Study

Description of Study

The GARNET trial (Study 4010-01-001)18 is an ongoing nonrandomized, noncomparative, multi-centre, open-label, phase I dose-escalation and cohort-expansion study in patients with recurrent or advanced solid tumours. The objective of part 2B of the GARNET trial was to evaluate the safety and antitumour activity of dostarlimab in patients with advanced solid tumours. Cohort A1 included patients with advanced or recurrent dMMR or MSI-H EC that had progressed on or after prior treatment with a platinum-containing regimen. Patients were enrolled from 123 sites in 8 countries (including 8 Canadian sites). Enrolment started on April 10, 2017, and is ongoing.

To be eligible, patients had to be at least 18 years of age, diagnosed with recurrent or advanced dMMR or MSI-H EC, and had to have progressed on or after no more than 2 lines of prior systemic therapy, with at least 1 of these being platinum-based doublet therapy. In addition, patients had to have adequate organ function and an Eastern Cooperative Oncology Group (ECOG) performance status of less than 1. The study consisted of 3 phases: the screening phase (up to 35 days before treatment), the treatment phase, and the follow-up phase. A total of 129 patients were enrolled in cohort A1. All patients received dostarlimab by IV injection (500 mg every 3 weeks for cycles 1 to 4, and 1,000 mg every 6 weeks from cycle 5 onward) for up to 2 years or until disease progression, treatment discontinuation, or withdrawal.

The co-primary outcomes of the GARNET trial were objective response rate (ORR) and duration of response (DOR). The secondary outcomes were OS, disease control rate (DCR), immune-related DCR (irDCR), PFS, immune-related PFS (irPFS), immune-related ORR (irORR), and immune-related DOR (irDOR). HRQoL was an exploratory outcome assessed by the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30) and the EQ-5D 5-Levels (EQ-5D-5L). The safety outcomes assessed included treatment-emergent adverse events (TEAEs), serious adverse events (SAEs), immune-related adverse events (irAEs), ECOG Performance Status, clinical laboratory measures, vital signs, electrocardiogram, physical examination, serum pregnancy testing, and concomitant medications.

The GARNET trial has 3 planned interim analyses that are contingent on a combined enrolment of 100, 200, and 300 patients, respectively, in cohort A1 and cohort F and 24 weeks of follow-up. Only data from the second interim analysis (IA-2) was reviewed by CADTH; it included a subset of patients from the first interim analysis (IA-1) (N = 72). Data for this subset of patients is available in Appendix 4. The data cut-off dates were July 8, 2019, March 1, 2020, and November 1, 2021, for IA-1, IA-2 (N = 105), and the third interim analysis (IA-3) (N = 143), respectively. The GARNET trial was sponsored by GlaxoSmithKline Inc.

Because only 2 (< 2%) enrolled patients had mismatch repair (MMR)-unknown but MSI-H tumours (MMR-unk/MSI-H), these patients were included with patients with dMMR tumours. Median age was 64 years (range = 39 to 80 years), median weight was 71 kg (range = 34.0 to 141.4), and median body mass index (BMI) was 27.97 kg/m2 (range = 13.6 to 53.9 kg/m2). Most patients were White (> 75%). The most common histology type of EC was type I endometrioid carcinoma (67.6%), with grade 2 being the most common histology grade at diagnosis (39%). More than two-thirds (67.6%) of patients had stage IV EC. ECOG Performance Status of 1 was the most common (60%), followed by ECOG 0 (40%). All patients received prior anti-cancer treatment. Most patients (88.6%) had 2 or fewer lines of prior anti-cancer regimens, and a smaller proportion had 2 or more lines (11.5%). More than half (56.2%) of patients had received prior regimens for metastatic disease. The patient subgroups of interest, as identified in the CADTH systematic review protocol, included the following: International Federation of Gynecology and Obstetrics (FIGO) stage, histology of tumour type (e.g., type I, type II) and subtype (e.g., clear cell carcinoma), number and type of prior systemic therapies (e.g., chemotherapy, hormonal therapy), prior radiation, and progression-free interval after the most recent platinum-containing anti-cancer therapy.

Outcomes

The key outcomes from cohort A1 of the GARNET trial are summarized in Table 2. At the time of IA-2, the median duration of follow-up was 16.3 months, and the median duration of treatment (DoT) was 26 weeks.

The proportion of patients who achieved an ORR (complete response [CR] or partial response [PR]) was 44.8% (95% confidence interval [CI], 35.0 to 54.8). The Best overall response (BOR) was CR in 11 patients (10.5%), PR in 36 patients (34.3%), and stable disease in 13 patients (37.1%). The DCR was 57.1% (95% CI, 35.0 to 54.8). Of those who responded, 89.5% had an ongoing response. The median DOR was not reached, but 79% of patients who achieved an objective response had a DOR of at least 6 months.

At the time of IA-2, a large proportion of patients (66.7% and 45.7%, respectively) had no OS or PFS events. The median OS was not reached, but Kaplan-Meier (KM) estimates for the probability of survival at 6, 9, and 12 months were 80.9% (95% CI, 71.7 to 87.4), 75.1% (95% CI, 65.2 to 82.6), and 68.9% (95% CI, 58.3 to 77.4), respectively. The median PFS was 5.5 months (95% CI, 3.2 to not reached), with KM estimates of PFS by RECIST 1.1 of 48.6% (95% CI, 38.6 to 57.9) at month 6 and 47.5% (95% CI, 37.4 to 56.8) at month 9 and at month 12. In terms of HRQoL, the EQ-5D-5L visual analogue scale (EQ-VAS) and EORTC QLQ-C30 scores appeared stable over time. Summary data for the EQ-5D-5L descriptive system were not provided.

As part of the sponsor’s feedback on this CADTH reimbursement review report, the sponsor provided CADTH with a summary of the updated analysis (data cut-off November 1, 2021) for certain baseline characteristics, efficacy, and safety outcomes in the GARNET trial. The results of the updated analysis were, overall, consistent with those reported in the previous analyses performed as of March 1, 2020, data cut-off date. The additional results from the November 1, 2021, data cut-off date are available in Appendix 5.

Harms Results

A summary of the key harms reported in cohort A1 of GARNET are summarized in Table 2. Almost all patients (95.3%) experienced at least 1 TEAE. The most common serious TEAEs were abdominal pain, acute kidney injury, sepsis, pulmonary embolism, pyrexia, and urinary tract infection. Grade 3 or higher TEAEs occurred in 48.1% of patients, with the most common being anemia (14.7%), abdominal pain (5.4%), and hyponatremia (3.9%). No patient withdrew due to an adverse event (AE) as a primary reason. Study treatment discontinuation due to AEs occurred in 11.6% of patients, whereas AEs that led to study treatment interruption occurred in 24% of patients. The most common AEs leading to study interruption were anemia (3.1%) and diarrhea (2.3%).

One patient died due to a TEAE (aspiration) during the treatment period, and 4 patients died due to TEAEs (i.e., pleural effusion, pneumonia, sepsis, and shock) during the 90-day safety follow-up. None of the TEAEs leading to death were considered treatment-related, and no TEAEs were the primary cause of death during the long-term follow-up period.

The notable harms associated with dostarlimab included immune-related toxicity. The incidence of irAEs was 34.9% in cohort A1. The most frequently reported irAEs (≥ 5%) were diarrhea and hypothyroidism. A total of 7.9% of patients had a serious irAE, 12.7% had an irAE of grade 3 or higher, and 4.8% had an irAE that led to study treatment discontinuation. Most of the irAEs were considered related to the study treatment.

Table 2. Summary of Key Results From the Pivotal Study (IA-2).

Table 2

Summary of Key Results From the Pivotal Study (IA-2).

Critical Appraisal

Internal Validity

The main limitation of the GARNET trial is the single-arm design, which makes it challenging to interpret the data and determine whether the efficacy and safety events observed were attributable to dostarlimab. Formal hypothesis and statistical significance testing were not performed, limiting the ability to draw conclusions. Given that results were based on an interim analysis, some time-to-event outcomes, including median OS and DOR, were not reached due to data immaturity; therefore, the treatment effect observed with dostarlimab may be overestimated. The risk of overestimating HRQoL benefit and known subjective harms is also high, given the open-label trial design in which treatment was not blinded. To mitigate bias, the sponsor used a blind independent clinical review (BICR) to evaluate treatment response, with standardized criteria for certain efficacy outcomes (i.e., ORR, DOR, PFS, and DCR). Therefore, bias is less of a concern for these end points and OS, and more of a concern for subjective end points, including HRQoL and safety. It is also acknowledged that mature OS data will be confounded by the use of subsequent anti-cancer therapy received by some patients after progressive disease. No analyses were undertaken to account for the potential of confounding. Overall, the magnitude and direction of bias is unclear. The clinical experts agreed that in the absence of robust comparative data on PFS and OS, no firm conclusions could be drawn on how dostarlimab compares with other relevant treatment options, as causal inferences cannot be made from the results of a single-arm trial design.

HRQoL was identified as an important outcome by the patient and clinician groups providing input for this review. However, no conclusions could be drawn from the HRQoL data from the GARNET trial due to several limitations. Given the wide and overlapping CIs, the reduced number of patient responses over time, and the lack of statistical testing and a definition of what constituted a clinically meaningful response, it is not possible to draw conclusions with precision from the available data.

External Validity

Overall, the clinical experts consulted by CADTH agreed that the inclusion and exclusion criteria, baseline patient characteristics, concomitant medications, and prohibited medications present in cohort A1 of the GARNET trial were reflective of patients they see in clinical practice for the indication under review. There were no barriers to identifying patients who would most benefit from the treatment, given that testing for MMR and microsatellite instability (MSI) status is standard practice in Canada. The clinical experts indicated that no difference in treatment effect would be expected based on variation in disease-management practices across participating countries. In the opinion of the clinical experts, as long as patients have dMMR or MSI-H tumour status, dostarlimab would be appropriate to administer after any of the prior therapies received by patients in the trial. However, they noted that clinical benefit may be diminished in patients with more prior lines of systemic therapy. There were a limited number of patients included in the primary efficacy analysis dataset (n = 105) and very few patients from various ethnic backgrounds, which may reduce the generalizability of the results to a real-world practice setting. Furthermore, the subgroup analyses had no statistical comparisons and even smaller sample sizes, which limits generalizability to a broader population.

Indirect Comparisons

Description of Studies

The sponsor submitted 6 reports of indirect treatment comparisons (ITCs) — 3 reports of matching-adjusted indirect comparisons (MAICs), and 3 reports of inverse probability of treatment weighting (IPTW) analyses19 — which aimed to compare survival between dostarlimab from the phase I GARNET trial with the current treatment paradigm in advanced or recurrent EC.

Efficacy Results

The primary end point for all comparisons was OS. Other outcomes included PFS, ORR, DOR, time to treatment discontinuation (TTD), DoT, time to next treatment (TTNT), time to deterioration in HRQoL, and AEs; however, these were less frequently investigated, and outcomes specifically important to patients, including HRQoL, were not assessed. The results of the MAIC and IPTW analyses generally suggest that dostarlimab is favoured for OS over all the included comparators.

Harms Results

The sponsor-submitted MAIC and IPTW reports did not assess safety outcomes.

Critical Appraisal

Although the results of the MAIC and IPTW analyses generally suggest that dostarlimab is favoured for OS over all the included comparators, there was significant uncertainty in the results based on the clinical heterogeneity of the included populations, resulting in reduced sample sizes and wide CIs. There were important differences in the design of the comparator studies that limit the ability to draw strong conclusions about the effectiveness for dostarlimab compared with other treatments. An important limitation of all analyses was the fact that MMR and MSI-H status was unknown for all or most patients in the comparator trial, and it is therefore uncertain whether the comparator population in the ITC analyses would be eligible for treatment with dostarlimab, providing further uncertainty about the comparative effectiveness.

Conclusions

One phase I, singe-arm, open-label trial (GARNET)18 provided evidence of the efficacy and safety of dostarlimab in adults with dMMR or MSI-H recurrent or advanced EC (cohort A1) that had progressed on or after prior treatment with a platinum-containing regimen. The clinical experts consulted by CADTH felt that the response outcomes (ORR and DOR co-primary outcomes) observed in the trial were clinically meaningful and durable for this patient population and, in their opinion, were higher than what is observed with currently used second-line therapies in this setting. The trial results were based on an interim analysis; therefore, there is the possibility of overestimating clinical benefit and underestimating harms. There was uncertainty around the magnitude of the clinical benefit, given the limitations inherent in the single-arm trial design. The trial data on important long-term outcomes were immature, and interpretation of OS will be confounded by the use of subsequent anti-cancer therapies. The clinical experts noted that a randomized controlled trial (RCT) would be needed to directly compare dostarlimab with currently available therapies in the second-line setting to accurately evaluate its efficacy in this patient population. In the absence of a direct comparison of dostarlimab with relevant treatment options, the sponsor submitted multiple ITCs. However, the CADTH critical appraisal of these analyses identified significant limitations with the submitted MAICs and IPTWs, which restricted the ability to interpret the relative treatment-effect estimates obtained. Limitations of the ITCs included heterogeneity across study designs, high risk of confounding and effect modifiers, and uncertainty regarding the inclusion of dMMR or MSI-H status in the comparator groups. The results for HRQoL, an outcome important to patients and clinicians, remained inconclusive due to the lack of statistical analysis, the substantial decline in patients completing questionnaires over time, and the lack of a definition of what constituted a clinical meaningful change from baseline. The notable harms observed with dostarlimab, such as diarrhea and peripheral nephropathy, were considered manageable and consistent with other immunotherapies by the clinical experts and, in their opinion, appeared favourable when naively compared with currently available chemotherapy options. However, interpreting the safety events attributable to dostarlimab was challenging because all patients in cohort A1 received the same treatment. Overall, limitations of GARNET’s single-arm design prohibited the drawing of causal conclusions between the intervention and outcomes.

Introduction

Disease Background

EC is the most common gynecological malignancy among women in Canada.1 EC malignant tumours arise from the cells of the uterine lining. More than 95% of all uterine cancers are endometrial.20 Uterine cancer is the 17th leading cause of cancer death in Canada.3,20 The Canadian Cancer Society estimated that 8,000 women would be diagnosed with uterine cancer in 2021 and that 1,400 women would die of the disease.2,3 EC most often occurs in patients older than 50 years, with an average age of diagnosis at 60 years.21 Diagnosis of EC occurs at an early stage for approximately 80% of patients because of the early presenting symptom of uterine bleeding.1,20 The most common route of diagnosis of EC is endometrial biopsy, followed by endometrial curettage and hysterectomy specimen.1 EC uses the FIGO criteria to determine disease stage, which depends on the size of the tumour and the extent to which the tumour has spread to lymph nodes or distant sites (metastasis).22 Generally, the higher the stage number, the more the cancer has spread.23 Tumour stage is fixed, regardless of tumour type.1

The prognosis of EC is primarily based on stage of cancer, histology of the tumour, and grade. Five-year survival by FIGO stage is 80% to 90% for stage I, 70% to 80% for stage II, and 20% to 60% for stage III and IV.1 In terms of histology, there are 2 subtypes: type I ECs represent 80% of patients and are low-grade (1 or 2) endometrioid tumours,1,20,24 and type II accounts for 10% to 20% of ECs and includes grade 3 endometrioid tumours and tumours of nonendometrioid histology, such as serous clear cell, mucinous, squamous, transitional cell, mesonephric carcinosarcoma, and undifferentiated.1,20 The 5-year survival of type I is around 80% to 90%, while 5-year survival of type II EC is as low as 20%. Other notable prognostic factors for EC include race, age, uterine tumour location, peritoneal cytology results, and lymphovascular space invasion.1

Molecular testing of biomarkers during endometrial biopsy assists in the identification of treatment options and in risk stratification.1 Standard testing includes immunohistochemistry (IHC) and polymerase chain reaction. IHC is used to test for dMMR, in which the cells’ ability to repair mistakes during the division process is impaired. The tumour is immunohistochemically assessed for the loss of at least 1 of the following MMR proteins: MLH1, MSH2, MSH6, and/or PMS2.25 The dMMR proteins cause cellular hypermutations and high levels of microsatellite instability (MSI-H) in sections of DNA. If MMR status cannot be determined from tumour samples, the sample may undergo genetic testing that uses next-generation sequencing (NGS) to identify MSI status. dMMR and MSI-H tumour statuses are predictive of clinical benefit from PD-1 inhibitors and represent approximately 25% of primary ECs and 13% to 30% of recurrent ECs.7-9 Recurrence occurs in approximately 13% to 20% of patients with EC, with rates varying greatly by FIGO stage at diagnosis; rates are highest among patients with stage IV EC (> 65%).4,5,26 The prognosis of patients with recurrent EC is poor, with a median survival of approximately 12 months.4

Standards of Therapy

Treatment options for EC are dependent on stage and pathologic factors identified after initial surgery and are based on estimated risk of disease recurrence. Early-stage EC and/or type I tumour ECs can be cured with surgery alone.5,27,28 Individuals diagnosed with advanced or recurrent EC may require adjuvant radiotherapy and/or chemotherapy, depending on the extent and location of spread and/or pathologic risk factors. CCO and Alberta Health Services recommendations favour combination chemotherapy over single-drug chemotherapy for individuals with advanced or recurrent EC, as combination therapy elicits a higher response rate while maintaining acceptable toxicity levels.5,29-32 The current standard of care for patients with advanced or recurrent disease is platinum-based chemotherapy as a doublet or single drug.29 A standard echoed by the European Society of Gynaecological Oncology, the European Society of Radiotherapy and Oncology, and the European Society of Pathology.33 The most common platinum-based therapy is carboplatin plus paclitaxel.5,29,33 For a subset of patients with low-grade recurrent or metastatic EC who are estrogen- or progesterone-receptor positive or for patients with poor tolerance to systemic therapy, hormonal therapy, such as megestrol, letrozole, and medroxyprogesterone, may be used.29

For patients with newly diagnosed advanced or recurrent EC, response rates for standard first-line treatment range from 40% to 62%.10-13 However, for patients with advanced or recurrent EC who have progressed on or after platinum-based chemotherapy, there is currently no standard effective or curative second-line therapy.5,33 Patients with recurrent EC are typically re-treated with platinum-based chemotherapy with poor outcomes; response rates range from 10% to 15% for all available treatment options.33 The clinical experts consulted by CADTH noted that median survival ranges from 12 to 15 months after re-treatment. Various single-drug chemotherapies may be administered to patients who are resistant or refractory to platinum-based chemotherapy, with response rates typically below 15% and no known clear survival benefit.14 Hormonal treatments may also be used for disease control but are not considered curative.

The clinical experts consulted by CADTH for this review indicated that there is great unmet need for effective therapies with acceptable toxicity profiles that achieve disease control, reduce disease-related symptoms, improve HRQoL, prevent disease progression, and prolong survival among patients with recurrent or advanced EC that has progressed on or after prior treatment with a platinum-containing regimen. There is currently no standard effective second-line therapy for recurrent or refractory disease, and commonly used therapies are noncurative. The clinical experts anticipated more promising benefit with biomarker-driven treatments for patients with dMMR or MSI-H cancers.

Drug

Dostarlimab is an anti-PD-1 monoclonal antibody that targets the cellular pathway between the PD-1 receptor and 2 programmed death ligands, PD-L1 and PD-L2, found on immune cells. Dostarlimab binds to the PD-1 receptor and blocks its interaction with PD-L1 and PD-L2. This suppresses the PD-1-pathway-mediated immune response in the tumour microenvironment, enhancing a patient’s antitumour immune response.15

On December 23, 2021, dostarlimab was issued a Notice of Compliance with conditions (NOC/c) by Health Canada for the treatment of adults with dMMR or MSI-H recurrent or advanced EC that has progressed on or after prior treatment with a platinum-containing regimen. The sponsor’s requested reimbursement criteria for dostarlimab align with the Health Canada indication. Dostarlimab underwent an expedited Health Canada review process (advance consideration under NOC/c); the market authorization comes with conditions, pending the results of trials that confirm its clinical benefit. Dostarlimab has no other Health Canada–approved indications and has not previously been reviewed by CADTH.

Dostarlimab received accelerated approval from the FDA in August 2021 for the treatment of adults with dMMR or MSI-H recurrent or advanced EC that has progressed on or after prior treatment with a platinum-containing regimen.34 Dostarlimab received conditional authorization in the European Union in April 2021 for the same indication.35 Currently, dostarlimab is being reviewed by NICE and the Scottish Medicines Consortium.16,17

Dostarlimab is available as a 500 mg IV infusion and is administered with an IV infusion pump over 30 minutes.15 The recommended dosage of dostarlimab as monotherapy in adults is 500 mg once every 3 weeks for dose 1 through 4, and 1,000 mg once every 6 weeks for dose 5 onward (dose 5 occurs 3 weeks after dose 4). The product monograph states that treatment may continue until disease progression or unacceptable toxicity. Dose reductions of dostarlimab are not recommended, but dosing delays and discontinuation are permitted based on safety and the patient’s tolerability of the treatment. Patients should be selected for treatment based on MSI-H or dMMR tumour status, determined by an accredited laboratory using validated testing methods.

Key characteristics commonly used in the treatment of advanced or recurrent dMMR or MSI-H EC are presented in Table 3.

Table 3. Key Characteristics of Dostarlimab, Platinum-Based Therapy, and Hormonal Therapy Regimens.

Table 3

Key Characteristics of Dostarlimab, Platinum-Based Therapy, and Hormonal Therapy Regimens.

Stakeholder Perspectives

Patient Group Input

The information in this section is a summary of input provided by the patient groups that responded to CADTH’s call for patient input for the purpose of this CADTH review. The full patient input received is available in the Stakeholder Input section.

The patient and caregiver input received for this review was collected by the Canadian Cancer Society. The input was sourced from on an online survey and from patient and caregiver testimonials gathered from October 22 to November 3, 2021. Six testimonials and 22 survey responses were received (20 patients with current or previous EC, and 2 caregivers of someone with current or previous EC). Of the 22 survey respondents, 8 (6 patients and 2 caregivers) had experience with dostarlimab. Six of these respondents resided in Quebec and 2 resided in British Columbia. All patients and caregivers who had experience with dostarlimab reported receiving it through a clinical trial.

Respondents indicated a range of EC symptoms that affected their daily activities. The daily activities that were most commonly moderately or severely affected included the ability to preform household chores (46%), travel (41%), exercise (41%), work (36%), fulfill family obligations (32%) and spend time with family and friends (27%). Forty-four percent of patients were not currently being treated for EC and 39% were undergoing immunotherapy. The treatment side effects most commonly reported as having a moderate or severe impact on daily life were issues with libido and sexual function (45%) and fatigue (41%). Fifty-nine percent of patients reported a financial barrier related to their treatment; among these patients, a loss of income due to absence from work (31%) and travel costs for cancer treatment (31%) were the most common barriers.

According to the patient input received, respondents expect the following key outcomes to be improved with any new drug or treatment: quality of life, periods of remission, drug affordability, access across jurisdictions, and fewer side effects (such as skin issues, fatigue, bladder control, stamina, hair loss, pain, arthritis, vaginal dryness, vaginal bleeding after intercourse, and concentration problems). All survey respondents indicated that, compared with other therapies, dostarlimab was easier to use either because it had little to no side effects (75%), longer intervals between doses (13%), or a shorter infusion time (13%).

Clinician Input

Input From Clinical Experts Consulted by CADTH

All CADTH review teams include at least 1 clinical specialist with expertise in 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). The following input was provided by 2 clinical specialists with expertise in the diagnosis and management of EC.

Unmet Needs

Currently, there is no standard second-line therapy for individuals with recurrent EC who have progressed on or after prior treatment with a platinum-containing regimen. This represents a critical unmet need in this patient population. Both clinical experts agreed that biomarker-driven treatments are needed to guide selection of the most effective and durable treatment option.

Place in Therapy

Currently, patients with advanced or recurrent EC receive carboplatin plus paclitaxel (another platinum chemotherapy doublet) or a single drug as first-line therapy. For a subset of patients with estrogen- or progesterone-receptor-positive indolent low-grade metastatic or recurrent EC, hormonal treatments, such as megestrol, medroxyprogesterone, letrozole, and tamoxifen, are considered. For those who have progressed on platinum chemotherapy and are considered to have platinum-resistant or refractory disease, second-line therapy with single-drug chemotherapy is considered, but it has a low expected response rate and a short DOR with no known survival benefit. Hormonal treatments may also be used for disease control but are not considered curative.

Dostarlimab will likely cause a shift in the current treatment paradigm for patients with dMMR or MSI-H metastatic or recurrent EC. The mechanism of action of dostarlimab would address the underlying disease-specific process and biomarkers for patients with dMMR or MSI-H EC. The clinical experts felt it would be preferable to initiate treatment with dostarlimab before other therapies.

Patient Population

The clinical experts agreed that patients with dMMR or MSI-H EC tumours would most benefit from an immune checkpoint inhibitor. One clinical expert added that patients with dMMR or MSI-H metastatic or recurrent EC of any histology would benefit from dostarlimab, regardless of symptoms or prior treatment; however, the other clinical expert noted that a smaller magnitude of benefit may be observed with increased previous lines of systemic therapy.

The clinical experts mentioned that the diagnosis of EC typically relies on biopsy, which may be conducted in clinic. If technical issues make it difficult to obtain samples from the endometrial lining during an office biopsy, the procedure can be performed in the operating room using dilatation and curettage. IHC testing for MMR status is relatively inexpensive and considered standard practice. If the MMR status of a sample is unknown, it may be further analyzed with genomic testing to determine MSI status. A valid test would involve NGS.

The clinical experts noted that treatment with dostarlimab would not be suitable for patients any of the following characteristics:

  • very poor performance status
  • history of severe autoimmune disease
  • prior therapy with anti-PD-1, anti-PD-L1, or anti-PD-L2 drugs
  • known uncontrolled nervous system metastases and/or carcinomatous meningitis
  • poor medical risk due to serious uncontrolled medical disorder
  • nonmalignant systemic disease or active infection requiring systemic therapy
  • history of immunosuppression
  • MSS EC.
Assessing Response to Treatment

According to the clinical experts, the most important goals of treatment for recurrent or advanced EC would be to improve OS and PFS; reduce symptoms of cancer; improve functional status (i.e., ability to perform activities of daily living); improve HRQoL; and reduce the burden of disease on patients and caregivers. One clinical expert noted that many patients with advanced or recurrent EC suffer from pelvic symptoms (e.g., unresectable disease in the pelvis causing bleeding or pain), lung symptoms (e.g., dyspnea from metastases), and neurologic symptoms (e.g., brain metastases) or bone symptoms (e.g., painful bony metastases), and that alleviation of these symptoms would be a benefit of treatment.

In terms of a clinically meaningful response, the clinical experts recommended that in addition to clinical assessment of disease symptoms and duration of disease control, the use of standard irRECIST39 for the assessment of response to immunotherapeutic treatments is useful. One clinical expert noted that the Common Terminology Criteria for Adverse Events40 tool for CT imaging can be used to assess response to treatment. The same clinical expert also suggested that cancer antigen 125 (nonspecific) may be used to assess treatment response in addition to other methods, although it is not commonly used in EC. In terms of the timing of assessments, the clinical experts recommend that response to treatment should be assessed radiologically every 3 months, with blood work every month and clinical assessments every 2 to 3 months.

Discontinuing Treatment

According to the clinical experts, treatment with dostarlimab should be discontinued when there are radiological and clinical signs and symptoms of disease progression, treatment toxicities (e.g., grade 3 or higher adverse reactions), or intolerability to treatment.

Prescribing Conditions

According to the clinical experts consulted by CADTH, the diagnosis, treatment, and monitoring of patients with EC should be undertaken by a specialist, namely a gynecologist oncologist, medical oncologist, and/or surgeon. Biomarker testing to identify dMMR or MSI-H status is also recommended.

Clinical experts recommend that dostarlimab be administered in a hospital clinic that has multidisciplinary medical supports to manage potential immune-related side effects. The clinical experts noted that treatment may also be administered in outpatient clinics.

Additional Considerations

The clinical experts noted that re-treatment with dostarlimab is possible if patients experience recurrence, as long as they had no signs of toxicity or intolerability while using the drug. If a patient completed their treatment and achieved a durable response for up to 2 years, and a significant time period elapsed before they progressed, then re-treatment with dostarlimab could be considered. The clinical experts noted that 50% of patients will have disease recurrence within 1 year of completing treatment for recurrent EC and will need to restart treatment.

Clinician Group Input

This section was prepared by CADTH staff based on the input provided by clinician groups.

The information in this section is a summary of 7 inputs provided by the registered-clinician groups that responded to CADTH’s call for clinician input for the purpose of this CADTH review. The full clinician group inputs received are available in the Stakeholder Input section. Input was received from the following clinical groups:

  • British Columbia Cancer Provincial Gynecological Oncology Tumour Group
  • MUHC, Division of Gynecologic Oncology
  • OH-CCO Gynecological Drug Advisory Committee
  • PMCC, Gynecologic Cancers Disease Site Group, Medical Oncology Group
  • SCA
  • GOC
  • SBHSC, Division of Gynecologic Oncology.
Unmet Needs

The views of the clinician groups were overall consistent with the clinical experts consulted by CADTH, indicating that the most important treatment goals for advanced or recurrent dMMR or MSI-H EC are disease control, prolonged survival, delayed worsening of symptoms, maintenance of HRQoL, delayed disease progression, and an acceptable safety profile. Treatment for individuals diagnosed with recurrent EC is a critical unmet need. All clinician groups noted that although the prognosis for patients diagnosed with early-stage disease is good, for those with recurrent or metastatic EC, median OS is short. In fact, there is no effective second-line treatment for Canadians with EC. Response rates to second-line cytotoxic drugs are low (< 20%), with a median PFS of 3 to 4 months.

All clinician groups also noted that although Health Canada recently issued a NOC/c approval for pembrolizumab immunotherapy for dMMR or MSI-H EC, there is no funded access to this treatment. The lack of funding has created disparity in access to immunotherapy, as only those with insurance coverage or the capacity to self-pay can access pembrolizumab. Moreover, PMCC added that there is currently no patient-supported or compassionate-access program for immune checkpoint inhibitor therapies. The MUHC and SBHSC noted that the side-effect profile of combination therapy with pembrolizumab and lenvatinib was significant, with 66.9% of patients experiencing a grade 3 or 4 toxicity. The MUHC added that prolonging poor HRQoL with interventions associated with significant toxicity serves very little purpose.

Place in Therapy

All clinician groups indicated that all patients with advanced or recurrent EC would benefit from effective immunotherapy, but patients with MSI-H or dMMR subtypes would most benefit from treatment with an immune checkpoint inhibitor. All groups recommended that patients diagnosed with advanced or recurrent EC should be offered platinum-based chemotherapy as first-line therapy. However, the British Columbia Cancer Provincial Gynecological Oncology Tumour Group did acknowledge that treatment with immune checkpoint inhibitors may be an appropriate first-line therapy for patients who either refuse chemotherapy or for whom chemotherapy may be contraindicated or known to be poorly tolerated, provided dMMR or MSI-H status is confirmed. The MUHC further stated, “it is false economy to delay starting a highly effective treatment [dostarlimab] with minimal toxicity in this niche population.” The MUHC stated that trying second-line chemotherapy alone in this patient population is not advisable due to toxicity and lack of benefit.

MUHC indicated that when EC recurs in patients who initially responded well and dostarlimab is interrupted for any other reason, restarting treatment with dostarlimab would be appropriate. If patients progress on dostarlimab or other immunotherapies, palliative and supportive care options are considered.

Patient Population

Currently, the standard of care in Canada includes universal molecular characterization of all endometrial carcinomas for dMMR. Thus, identifying EC subtypes should not be a barrier to treatment. The British Columbia Cancer Provincial Gynecological Oncology Tumour Group and the GOC noted that patients whose EC is related to Lynch syndrome would benefit from treatment with dostarlimab, and PMCC added that those with polymerase-epsilon-mutation would benefit as well. The SCA noted that patients with EC suitable for treatment with dostarlimab should be PD-L1-naive and should not have undergone more than 2 prior treatments with a platinum-based therapy.

All clinician groups noted that patients who do not have a dMMR or MSI-H profile and/or have contraindications to immune checkpoint inhibitors are not suitable for treatment with dostarlimab. PMCC and the GOC also indicated that patients with MMR-proficient EC would not be suitable to receive dostarlimab as monotherapy treatment.

SBHSC noted that the EC patient groups less suitable for treatment with dostarlimab would be those that met all the following criteria:

  • no previous treatment with platinum-based chemotherapy
  • no dMMR seen on IHC
  • poor medical risk due to a serious uncontrolled medical disorder
  • poor performance status (ECOG Performance Status score of at least 3)41
  • inadequate organ function
  • a known immunodeficiency or current use of systemic steroids or other immunosuppressant medications.
Assessing Response to Treatment

The British Columbia Cancer Provincial Gynecological Oncology Tumour Group, COG, OH-CCO, and SBHSC noted that improvement in symptoms and physical findings related to advanced or recurrent EC (e.g., pain, bleeding, shortness of breath) would be considered a clinically meaningful treatment outcome. OH-CCO and PMCC stated that durable disease control with no adverse effects on HRQoL is an ideal treatment goal in this population. The SBHSC specified that PFS of at least 6 months would be a clinically meaningful outcome in this patient population. In addition, OH-CCO listed reduced caregiver burden as a meaningful treatment outcome.

The British Columbia Cancer Provincial Gynecological Oncology Tumour Group and the GOC also listed the following outcomes as clinically meaningful responses to treatment:

  • maintenance or improvement of performance status and ability to perform activities of daily living
  • evidence of disease regression from imaging studies
  • disease stabilization (in patients with good baseline performance status and few disease-related symptoms).

OH-CCO and the GOC reported that standard clinical monitoring of therapy, including a physical examination, symptom review, and intermittent CT imaging, should be used to evaluate response to treatment. The MUHC recommended that after the first 3 cycles of dostarlimab, patients should be assessed for response to treatment with a CT scan. If there is no indication of progression and patients are well, the CT-monitoring interval may be spaced out to every 12 weeks. PMCC recommended that tumour assessment by CT scan or MRI be completed every 2 to 3 cycles (i.e., every 6 to 9 weeks). The British Columbia Cancer Provincial Gynecological Oncology Tumour Group and SBHSC stated that CT imaging should occur every 12 weeks, whereas the SCA suggested imaging every 3 to 6 months. The British Columbia Cancer Provincial Gynecological Oncology Tumour Group and SCA also added that laboratory values, as well as respiratory and pulmonary status, should be assessed to ensure that treatment response is occurring without toxicity.

Discontinuing Treatment

All clinician groups indicated that treatment with dostarlimab should be discontinued if patients experience disease progression or serious toxicities. SBHSC indicated that the following side effects warrant consideration of pausing or discontinuing dostarlimab:

  • grade 2 anemia
  • pneumonitis
  • grade 3 colitis
  • grade 2 asthenia
  • grade 3 myalgia
  • pemphigoid
  • grade 3 increase in transaminases.

PMCC and SBHSC noted that due to the nature of immunotherapies and the possibility of pseudo-progression, patients with progression of disease on the first CT scan after initiation of treatment, but with no other symptoms, should continue treatment until further imaging demonstrates progression of disease. Similarly, the MUHC adds that in cases where disease progression occurs after 3 cycles with dostarlimab, patients should receive follow-up in 6 weeks to rule out pseudo-progression and discontinue treatment if progression is confirmed.

Prescribing Conditions

The British Columbia Cancer Provincial Gynecological Oncology Tumour Group, MUHC, OH-CCO, GOC, and SCA explained that dostarlimab is suitable to be delivered in the community and in outpatient and specialty clinics. Conversely, PMCC and SBHSC suggested that dostarlimab be delivered at cancer centres by gynecologic or medical oncologists. Both groups recommend that dostarlimab be administered in a chemotherapy suite with appropriate supervision by an oncologist familiar with gynecologic cancers and the management of immune-related adverse effects. The British Columbia Cancer Provincial Gynecological Oncology Tumour Group and the GOC noted that patients should be under the care of a treating physician or nursing staff with experience monitoring and evaluating patients for possible toxicities that may be caused by immune checkpoint inhibitor therapy. The GOC stated that this should not be a barrier to treatment because such therapies are now routinely used by most oncologists. The MUHC notes that for patients who do not live close to a treatment centre (e.g., patients in rural, remote, or Indigenous communities), it is appropriate to work closely with local physicians to share the responsibility of patient care.

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. Implementation questions from the drug programs 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.

Table 4

Summary of Drug Plan Input and Clinical Expert Response.

Clinical Evidence

The clinical evidence included in the review of dostarlimab is presented in 2 sections. The first section, the systematic review, includes pivotal studies provided in the sponsor’s submission to CADTH and Health Canada, as well as studies that were selected according to the a priori CADTH protocol. The second section includes indirect evidence from the sponsor and indirect evidence selected from the literature that met the selection criteria specified in the CADTH review.

Systematic Review (Pivotal and Protocol-Selected Studies)

Objectives

To perform a systematic review of the beneficial and harmful effects of dostarlimab in 500 mg to 1,000 mg doses administered intravenously for the treatment of adults with dMMR or MSI-H advanced or recurrent EC that has progressed on or after prior treatment with a platinum-containing regimen.

Methods

Studies selected for inclusion in the systematic review included pivotal studies provided in the sponsor’s submission to CADTH and Health Canada, as well as those meeting the selection criteria presented in Table 5. The systematic review protocol was established before the granting of the NOC/c from Health Canada. Outcomes included in the CADTH review protocol reflect outcomes considered to be important to patients, clinicians, and drug plans.

Table 5. Inclusion Criteria for the Systematic Review.

Table 5

Inclusion Criteria for the Systematic Review.

The literature search for clinical studies was performed by an information specialist using a peer-reviewed search strategy, according to the PRESS Peer Review of Electronic Search Strategies checklist.42

Published literature was identified by searching the following bibliographic databases: MEDLINE All (1946–) via Ovid and Embase (1974–) via Ovid. All Ovid searches were run simultaneously as a multi-file search. Duplicates were removed using Ovid deduplication for multi-file searches, followed by manual deduplication in Endnote. The search strategy comprised both controlled vocabulary, such as the National Library of Medicine’s MeSH (Medical Subject Headings), and keywords. The main search concept was dostarlimab. Clinical trials registries were searched: the US National Institutes of Health’s clinicaltrials.gov, WHO’s International Clinical Trials Registry Platform (ICTRP) search portal, Health Canada’s Clinical Trials Database, and the European Union Clinical Trials Register.

No filters were applied to limit the retrieval by study type. Retrieval was not limited by publication date or by language. Conference abstracts were excluded from the search results. Refer to Appendix 1 for the detailed search strategies.

The initial search was completed on November 3, 2021. Regular alerts updated the search until the meeting of the CADTH pan-Canadian Oncology Drug Review Expert Committee (pERC) on March 9, 2022.

Grey literature (literature that is not commercially published) was identified by searching relevant websites from the Grey Matters: A Practical Tool For Searching Health-Related Grey Literature checklist.43 Included in this search were the websites of regulatory agencies (FDA and European Medicines Agency). Google was used to search for additional internet-based materials. Refer to Appendix 1 for more information on the grey literature search strategy.

These searches were supplemented with a review of bibliographies of key papers and contacts with appropriate experts. In addition, the manufacturer of the drug was contacted for information regarding unpublished studies. Two CADTH clinical reviewers independently selected studies for inclusion in the review based on titles and abstracts, according to the predetermined protocol. Full-text articles of all citations considered potentially relevant by at least 1 reviewer were acquired. Reviewers independently made the final selection of studies to be included in the review, and differences were resolved through discussion.

A focused literature search for network meta-analyses dealing with EC was run in MEDLINE All (1946–) on November 3, 2021. No limits were applied to the search.

Findings From the Literature

Two reports of 1 study18,44 were identified from the literature for inclusion in the systematic review (Figure 1); the study is summarized in Table 6. A list of excluded studies is presented in Appendix 2.

A total of 31 citations were identified in the literature search, of which 2 were considered potentially relevant reports; and 23 potentially relevant reports were identified from other sources. Of these, 3 full-text reports were retrieved for scrutiny. In total, 1 report of 1 study was included in the CADTH review.

Figure 1

Flow Diagram for Inclusion and Exclusion of Studies.

Table 6. Details of Included Study.

Table 6

Details of Included Study.

Description of Study

The GARNET trial (Study 4010-01-001)18 is an ongoing multi-centre, single-arm, open-label, phase I dose-escalation and cohort-expansion study of patients with recurrent or advanced solid tumours. The primary objective of part 2B of the GARNET trial was to evaluate the safety and antitumour activity of dostarlimab in patients with advanced solid tumours. The secondary objective was to characterize the pharmacokinetic profile and to evaluate immunogenicity and additional measures of clinical benefit of dostarlimab. Exploratory objectives were to characterize the pharmacodynamics of dostarlimab and patient-reported outcomes assessed with EQ-5D-5L and the EORTC QLQ-C30. Patients were assigned to different cohorts based on variety of factors, including cancer type, response to previous therapies, and/or clinical biomarkers. Cohort A1 included patients with recurrent or advanced dMMR or MSI-H EC who had progressed on or after prior treatment with a platinum-containing regimen. Cohort A2 included patients with recurrent or advanced MMR-proficient or MSS EC that had progressed on or after prior treatment with a platinum-containing regimen. Cohort F included patients with nonendometrial dMMR, MSI-H, or polymerase-epsilon-mutation cancers who had progressed after 2 prior lines of systematic therapy for recurrent or advanced disease and had no alternative treatment options. For the purpose of this review, only the results of cohort A1, which aligns with the Health Canada indication, are presented. Refer to Figure 2 for a schematic of the GARNET trial.

A total of 129 patients were enrolled into cohort A1. Enrolment started on April 10, 2017 and is ongoing. Patients were enrolled across 123 sites in 8 countries (refer to Table 6). Of the 123 sites, 8 were located in Canada. Refer to Figure 2 for a schematic of the GARNET trial. All patients received dostarlimab by IV injection (500 mg every 3 weeks for cycles 1 to 4, and 1,000 mg every 6 weeks from cycle 5 onward) for up to 2 years or until disease progression, treatment discontinuation, or study withdrawal. Patients were permitted to continue treatment with dostarlimab beyond 2 years if the treating physician and the sponsor agreed that the treatment continued to provide clinical benefit to the patient.

To be enrolled, patients had to have archival or newly obtained tumour tissue so the tumour microenvironment could be assessed for biomarkers; in cohort A1, this included IHC results from a certified local laboratory that could be used to determine dMMR status. The definition of dMMR was the loss of expression of 1 of the following proteins, determined with IHC testing: MLH1, MSH2, MSH6, and PMS2. In the absence of known MMR status, MSI-H was tested using NGS in a central laboratory. Assay results assessed with local polymerase chain reaction were also acceptable for the determination of MSI status.

The study consisted of 3 phases: the screening phase (up to 35 days before beginning treatment), the treatment phase, and the follow-up phase. During the screening phase, 479 patients were screened for enrolment in either cohort A1 or cohort A2, based on MMR/MSI status. There were 184 screening failures, and 5 patients were screened but not treated. During the follow-up phase, patients were followed for safety (final follow-up visit was 90 ± 7 days after the end of treatment [EOT]), as well as for OS and HRQoL. The date of the final analysis for OS had not been determined at the time of the data cut-off (March 1, 2020).

The GARNET trial has 3 planned interim analyses that are contingent on a combined enrolment of 100, 200, and 300 patients, respectively, in cohort A1 and cohort F. For IA-1, the data cut-off date was July 8, 2019, after a 100 patients with measurable disease at baseline and at least 24 weeks of follow-up had been enrolled in either cohort A1 or cohort F. For IA-2, the data cut-off date was March 1, 2020, after the combined patient enrolment of cohort A1 and cohort F reached approximately 200 patients with measurable disease at baseline and at least 24 weeks of follow-up. For IA-3, the data cut-off date was November 1, 2021, after the combined patient enrolment in cohort A1 and cohort F reached approximately 300 patients with measurable disease at baseline and at least 24 weeks of follow-up. Only data from the IA-2 data cut-off date were reviewed by CADTH, which included a subset of 72 patients from IA-1 (refer to Appendix 4 for further details). The GARNET trial was sponsored by GlaxoSmithKline Inc.

Amendments and Protocol Deviations

The study protocol of the GARNET trial was amended 6 times, although versions 4.0 and 5.0 were not implemented. Amendments to the protocol did not substantially affect the results of the study.

Amendment 2 (October 31, 2016) included updates to the inclusion criteria, tumour assessment criteria, definitions for safety data, and sample size justification of cohort A1 and cohort A2.

Amendment 3 (October 9, 2017) included the addition of HRQoL assessments and interim analyses for patients with dMMR or MSI-H cancer in cohort A1 and cohort F combined.

Amendment 4 (July 3, 2018) included revisions to allow clinically stable patients without major safety issues to continue with dostarlimab treatment after confirmation of progressive disease. Changes were also made to blood sample collection and to increase the number of patients in cohort A2.

Amendment 5 (May 10, 2019) implemented changes to the definition of cohort A1 and cohort A2, based on feedback from a regulatory agency. A recommendation was made to select patients based on the MMR IHC result (local or central test) instead of NGS; hence, the central testing vendor was changed. The sample size for cohort A1 was increased and the assessment of HRQoL was revised from a secondary to an exploratory objective.

Amendment 6 (January 7, 2020) updates to the primary objective included evaluation of the safety and tolerability of dostarlimab, as well as the addition of 2 interim analyses for the combined enrolment of cohort A1 and cohort F to reach 200 and 300 patients, respectively, and for all patients to have measurable disease at baseline and at least 24 weeks of follow-up.

The study design flow of the pivotal trial. In part 2B, patients were randomized into either cohort A1 or cohort A2 depending on MMR and MSI status.

Figure 2

Study Schema.

Populations

Inclusion and Exclusion Criteria

The key inclusion and exclusion criteria used in cohort A1 of the GARNET trial are summarized in Table 4. Briefly, the trial enrolled adults aged 18 years and older who had been diagnosed with recurrent or advanced EC and who progressed on or after no more than 2 lines of prior systemic therapy (not including hormone therapy), with at least 1 of these being platinum-based doublet therapy. Patients had to have documentation of dMMR or MSI-H status and radiologically measurable disease that met Response Evaluation Criteria in Solid Tumors 1.1 (RECIST 1.1).45 All EC histologies were permitted, except for endometrial sarcoma (including carcinosarcoma). At screening, patients had to have an ECOG Performance Status of 0 or 1, adequate organ function, a negative serum pregnancy test (if of childbearing potential), and 1 highly effective form of contraception throughout the study period. Prior therapy with an anti-PD-1, anti-PD-L1, or anti-PD-L2 drug was not permitted.

Baseline Characteristics

The baseline characteristics of patients who comprised the primary efficacy population of the GARNET trial are summarized in Table 7. At baseline, 105 patients in cohort A1 were identified as having dMMR or MSI-H EC (103 dMMR and 2 MMR-unk/MSI-H EC patients) for the primary efficacy analysis at IA-2. The demographic and baseline characteristics of patients with dMMR or MMR-unk/MSI-H tumours are presented as a total because of the small proportion of patients (n = 2) with MMR-unk/MSI-H tumours; the sponsor noted that the data remained similar when these patient groups were pooled. The median age was 64 years (range = 39 to 80 years), with 50.5% of patients between 39 and 65 years. Median weight was 71 kg (range = 34.0 to 141.4), and median BMI was 27.97 kg/m2 (range = 13.6 to 53.9 kg/m2). Most patients were White (78.1%). The most common histology type was type I endometrioid carcinoma (67.6%); the most common histology grade at diagnosis was grade 2 (39.0%). At the time of study enrolment, more than two-thirds (67.6%) of patients had FIGO stage IV EC. An ECOG Performance Status of 1 was the most common (60%), followed by an ECOG Performance Status of 0 (40%). All patients received prior anti-cancer treatment that included surgery (90.5%), radiotherapy (70.5%), adjuvant or neoadjuvant treatment (53.3%), and/or bevacizumab use (4.8%). Most patients (88.6%) had previously received 2 or fewer lines of anti-cancer regimens; a small portion had previously received 2 or more lines (11.5%). More than half (56.2%) of patients had received prior regimens for metastatic disease.

Table 7. Summary of Baseline Characteristics of Patients in the GARNET Trial (IA-2, Primary Efficacy Analysis Dataset) .

Table 7

Summary of Baseline Characteristics of Patients in the GARNET Trial (IA-2, Primary Efficacy Analysis Dataset) .

Interventions

All patients in cohort A1 received dostarlimab as a 30-minute IV infusion (with a permitted window of –5 minutes and + 15 minutes). Doses were administered in hospitals, infusion centres, and outpatient clinics. Dosing schedules were as follows:

  • cycles 1 to 4: 500 mg every 3 weeks (day 1 of each 21-day cycle)
  • cycle 5 and onward: 1,000 mg every 6 weeks (day 1 of each 42-day cycle).

Patients continued with the study treatment for up to 2 years or until specific withdrawal criteria were met. Patients discontinued study treatment for the following reasons: AEs, including dose-limiting toxicities; progressive disease based on clinical criteria used by the investigator or outlined in the protocol; patient request or patient pregnancy; risk to patients, determined by the investigator and/or sponsor; and severe noncompliance with the protocol, determined by the investigator and/or sponsor. In some instances, study treatment continued beyond 2 years if the treating physician and the sponsor agreed that treatment with dostarlimab continued to provide clinical benefit to the patient.

Dose Modifications (Delay, Interruption)

Dosing delays were permitted in the case of medical or surgical events or for logistical reasons not related to study treatment (e.g., surgery, unrelated medical events, participant vacation). Continuation of the study treatment should have occurred within 28 days of the scheduled dostarlimab infusion. If a delay was more than 28 days, a patient may have been permitted to continue treatment after discussion with the sponsor.

Dosing interruptions were permitted in the event of toxicities. In general, treatment withheld for drug-related grade 3 toxicities could be resumed if toxicity resolved to grade 1 or lower. Similarly, for all irAEs, dostarlimab was held until the patient was considered clinically and metabolically stable and AEs resolved to grade 1 or lower. Dostarlimab was permanently discontinued after any drug-related grade 4 events and after some grade 3 immunologic-mediated AEs.

Dose reductions were not permitted.

Concomitant Medications

Concomitant medications were allowed to treat AEs and comorbidities. Concomitant medications included any medications other than study treatments taken on or after the initial study treatment dosing date. If systemic steroids were used as a part of irAE management, the total dose of daily steroids allowed was 10 mg or less of prednisone when dostarlimab treatment was resumed. Patients were also allowed to receive rescue medications and appropriate supportive care deemed necessary by the treating investigator.

To ensure accurate assessment of the safety and efficacy of dostarlimab, patients were prohibited from receiving the following therapies during the screening and treatment periods of the study:

  • systemic anti-cancer or biological therapy
  • immunotherapy not specified in the protocol
  • chemotherapy not specified in the protocol
  • investigational drugs other than dostarlimab
  • radiation therapy within 3 weeks before study day 1 and during study treatment
    • note: palliative radiation therapy to a small field more than 1 week before day 1 of study treatment may have been allowed
  • any surgery that involved tumour lesions
    • note: radiation therapy or surgery that involved tumour lesions was considered to be progressive disease at the time the procedure was performed.
Subsequent Therapies

Data were collected on whether patients received subsequent anti-cancer therapy after treatment discontinuation or progressive disease. However, these were not defined in the protocol or statistical analysis plan.

Outcomes

A list of the efficacy end points identified in the CADTH review protocol that were assessed in the clinical trials included in this review is provided in Table 8. These end points are further summarized below. A more detailed discussion of the HRQoL and symptom severity outcome measures assessed in the trial is provided in Appendix 3.

Table 8. Summary of Outcomes of Interest Identified in the CADTH Review Protocol.

Table 8

Summary of Outcomes of Interest Identified in the CADTH Review Protocol.

Response assessment, according to RECIST 1.1,45 was based on radiologic imaging and performed by a BICR at pre-specified time points for the outcomes of OS, DCR, PFS, DOR, and BOR. The first radiographic evaluation occurred 12 weeks (84 ± 10 days) after the first dostarlimab dose and every 6 weeks (42 ± 10 days) thereafter, independent of cycle delays and/or dose interruptions. After 48 weeks of radiographic assessments, imaging and assessment of serum-based tumour markers were performed every 12 weeks (84 ± 10 days) until progressive disease. If a patient discontinued treatment for any reason other than progressive disease, death, withdrawal of consent, or loss to follow-up, radiographic scans and appropriate testing of serum-based tumour markers continued at the specified intervals until progressive disease was confirmed or an alternate anti-cancer therapy was started, whichever occurred first.

Tumour response was also evaluated by the investigator based on irRECIST 1.146,47 for the outcomes of immune-related DCR (irDCR), immune-related PFS (irPFS), immune-related ORR (irORR), and immune-related (irDOR). Investigators used irRECIST-based assessment to make clinical decisions. Per irRECIST, immune-related complete response (irCR) or immune-related partial response (irPR) was confirmed with repeat radiographic evaluation, at the earliest, 4 weeks after the first indication of response or at the next scheduled scan, whichever was clinically indicated. Progressive disease was confirmed with radiographic evaluation a minimum of 4 weeks and up to 6 weeks after the first progressive disease assessment.

Overall Survival

OS was a secondary outcome of the GARNET trial and was defined as the time from the first dose of study treatment to death by any cause. Patients last known to be alive were censored at the date of the last known contact, as follows: OS in days = date of death or censoring – date of first dose + 1.

OS was assessed at EOT, at the safety follow-up visit, and every 3 months after treatment.

Disease Control Rate and Immune-Related Disease Control Rate

DCR and irDCR were secondary outcomes of the GARNET trial. DCR, per RECIST 1.1., was defined as the proportion of patients achieving BOR of confirmed CR, PR, or stable disease. irDCR, per irRECIST 1.1., was defined as the proportion of patients achieving irBOR or irCR. irPR or immune-related stable disease as assessed by the investigator.

BOR was programmatically derived, based on reported time point response assessments determined by a central reader at different evaluation time points, from the first dose until the first documented progressive disease. The irBOR, according to irRECIST, was programmatically derived, based on reported overall time point responses determined by investigators after assessment of radiological scans, at different evaluation time points from the first dose until documented disease progression.

BOR was determined according to the following rules:

  • CR — at least 2 consecutive determinations of CR more than 4 weeks apart, with no other assessment between the 2 determinations other than not evaluable, CR, or missing.
  • PR — at least 2 consecutive determinations of PR or better more than 4 weeks apart, with no other assessment between the 2 determinations other than not evaluable, CR, PR, or missing before disease progression (and not qualifying for a CR).
  • Stable disease — at least 1 stable disease or non-CR or nonprogressive disease assessment (or better) at least 12 weeks to 10 days (i.e., ≥ 74 days) after baseline and before progressive disease (and not qualifying for a CR or PR). For an unconfirmed PR or CR to qualify, it must still meet the requirement of at least 12 weeks to 10 days.
  • Progressive disease — disease progression after baseline. Note that a determination of CR followed at least 4 weeks later by stable disease resulted in a BOR of progressive disease.
  • No disease — for central imaging data, when the independent radiologist cannot identify any disease at baseline, all subsequent assessments were documented as no disease if not declared progressive disease or not evaluable.

irBOR followed similar rules, except outcomes were immune-related (e.g., irCR, irPR). Clinical deterioration was not considered to be progressive disease. Only tumour assessments performed before the start of any new anti-cancer treatment were considered in the assessment of BOR and irBOR.

Progression-Free Survival and Immune-Related Progression-Free Survival

PFS and irPFS were secondary outcomes of the GARNET trial. PFS was defined as the time from the first dose to the earlier assessment of progressive disease or to death by any cause in the absence of progressive disease. irPFS time was defined, per irRECIST 1.1, as the time from the first dose to the earlier date of assessment of immune-related disease progression (irPD) event or to death by any cause in the absence of disease progression.

irPD events were considered when:

  • 2 consecutive irPDs were observed; the date of the first was considered to be the date of the irPD event.
  • the only or most recent tumour assessment before treatment discontinuation was irPD; the date was considered to be the date of the irPD event.

PFS and irPFS times were defined as follows: PFS or irPFS in days = date of PD or irPD event or death ÷ censoring – date of first dose + 1 (refer to Table 9 for details on censoring rules for PFS and irPFS).

Table 9. Censoring Rules for DOR, irDOR, PFS, irPFS.

Table 9

Censoring Rules for DOR, irDOR, PFS, irPFS.

Objective Response Rate and Immune-Related Objective Response Rate

ORR was the primary outcome of the GARNET trial and was defined, per RECIST 1.1 based on BICR, as the proportion of patients that achieved a BOR of CR or PR. irORR was a secondary outcome of the GARNET trial and was defined, per irRECIST 1.1, as the proportion of patients that achieved irBOR of irCR or irPR. Nonresponders were patients who did not have a post-baseline radiographic tumour assessment, who received post-baseline antitumour treatments (including surgery or radiation to tumour lesions) other than the study treatments before reaching a CR or irCR or a PR or irPR, or who died, progressed, or dropped out for any reason before reaching a CR or irCR or a PR or irPR.

Duration of Response and Immune-Related Duration of Response

DOR was a co-primary outcome of the GARNET trial and was defined as the time from first documentation of overall response leading to a confirmed CR or PR until the time of first documentation of disease progression or death. Censoring rules for DOR were similar to those for PFS (refer to Table 9 for details on censoring rules). irDOR was a secondary outcome of the GARNET trial and was defined, per irRECIST 1.1, as the time from first documentation of response leading to a confirmed irCR or irPR until the time of the irPD event or death. As with BOR and irBOR, clinical deterioration was not considered to be documented progressive disease.

Health-Related Quality of Life

The HRQoL outcomes measured in the trial included EORTC QLQ-C30 and EQ-5D-5L scores. Data from both instruments were exploratory outcomes in the GARNET trial and assessed at IA-2. HRQoL assessments were collected at scheduled visits (at each cycle) every 3 weeks (± 7 days) for the first 12 weeks, beginning on cycle 1, day 1, and every 6 weeks (± 7 days) thereafter while the patient was receiving study treatment. After treatment discontinuation, HRQoL assessments were collected from the remaining patients at the EOT visit, at the safety follow-up visit (90 days after EOT), and every 90 days (± 14 days) during the post-treatment follow-up period. A detailed discussion and critical appraisal of the HRQoL measures are provided in Appendix 3.

The EORTC QLQ-C30 is a questionnaire developed specifically to assess HRQoL in cancer patients. The questionnaire consists of 30 questions, 5 function scales (physical, role, cognitive, emotional, and social), 1 global health status/global quality-of-life scale, 3 symptom scales (fatigue, pain, and nausea and vomiting), and 6 single items that assess additional symptoms (dyspnea, appetite loss, sleep disturbance, constipation, and diarrhea) and financial impact.48 Scales and single items range in score from 0 to 100, with higher scores on the functional and global health status/quality-of-life scales indicating higher levels of functioning and health status/quality of life, respectively. Higher scores on symptom scales or items represent a greater presence of symptoms.48

The EQ-5D-5L version 2.0 is a generic HRQoL questionnaire for assessing a patient’s health status in terms of a single index value or utility score. There are 2 components of the questionnaire: a descriptive system that allows patients to rate their level of problems (none, slight, moderate, severe, extreme/unable) in 5 areas (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression), and a visual analogue scale that allows patients to rate their overall health status from 0 (worst imaginable) to 100 (best imaginable).49

Safety

Assessing the safety of dostarlimab was a primary objective of the GARNET trial. The primary safety variables included the following:

  • TEAEs — all, SAEs, their relationship to the study treatment, and their intensity (based on National Cancer Institute Common Terminology Criteria for Adverse Events v4.03 severity grades)
  • TEAEs leading to discontinuation
  • TEAEs leading to death
  • irAEs
  • clinical laboratory measures (including hematology, chemistry, coagulation, thyroid function, and urinalysis) and vital signs
  • ECG
  • physical examination
  • ECOG status
  • serum pregnancy testing
  • concomitant medications.

Statistical Analysis

Sample Size Determination

For the final analysis of the co-primary end point (ORR) in cohort A1, the sample size of approximately 100 patients with dMMR or MSI-H EC (with potential enrolment of up to 165 patients) was planned after protocol amendment 5. The null hypothesis that the true response rate was 20% or less (expected ORR for conventional therapy) was tested against a 1-sided alternative hypothesis of a response rate of at least 40%. With 65 patients in cohort A1, there was 92% power to rule out an ORR of 20% or less when the true ORR was 40% at the 2.5% type I error rate (1-sided). The increase in sample size allowed the lower-limit boundary of the exact 95% CI to exclude a response rate of 25% or less, assuming the observed ORR was 35%.

Interim Analyses

The 3 planned interim analyses were based on the combined enrolment in cohort A1 and cohort F (nonendometrial cancer). IA-1, IA-2, and IA-3 were conducted when the combined enrolment reached 100, 200, and 300 patients, respectively. All enrolled patients in cohort A1 and cohort F had dMMR tumours with measurable disease at baseline and at least 24 weeks of follow-up. IA-1 had a data cut-off date of July 8, 2019; IA-2 had a data cut-off date of March 1, 2020; and IA-3 had a data cut-off date of November 1, 2021. IA-2 and IA-3 were added after amendment 6.

All interim analyses evaluated the outcomes listed in Table 8, with the exception of IA-1, which included irORR as a primary end point and patient-reported outcomes as a secondary end point.

Primary Outcomes

The co-primary outcomes in the GARNET trial were ORR and DOR. No formal hypothesis-testing or inferential analyses were performed in cohort A1, and no statistical comparisons were planned between cohorts. The analysis of ORR included summary statistics, including the number of patients and percentage for categorical variables and, for continuous variables, the number of patients, mean, standard deviation (SD), median, minimum, and maximum. Two-sided exact 95% CIs based on the Clopper-Pearson method were reported for the ORR.

DOR analyses were performed using KM methods and summarized by minimum, maximum, 25th, 50th (median), and 75th percentiles with associated 95% CIs, the number and percentage of events, the number and percentage of censored observations, and the median duration of follow-up. Censoring rules for DOR are presented in Table 7.

Secondary Outcomes

The analyses for OS were based on the safety analysis and primary efficacy analysis datasets. OS analyses were performed using KM methods and summarized by the 25th, 50th (median), and 75th percentiles with associated 95% CIs, as well as the number and percentage of events and censored observations. Censoring for OS was set at the last known date of contact.

The analyses of DCR and irDCR involved summary statistics, including the number of patients and percentage for categorical variables and, for continuous variables, the number of patients, mean, SD, median, minimum, and maximum. Two-sided exact 95% CIs based on the Clopper-Pearson method were provided for DCR and irDCR.

The analyses of PFS and irPFS were performed using KM methods and summarized by the 25th, 50th (median), and 75th percentiles with associated 95% CIs, as well as the number and percentage of events and censored observations. Censoring rules for PFS and irPFS are presented in Table 9.

The analyses of irORR and irDOR were similar to the those of the co-primary outcomes of ORR and DOR.

Exploratory Outcomes

No analysis plan, objectives, or minimally important difference (MID) for the EORTC QLQ-C30 or the EQ-5D-5L instruments were specified a priori in the statistical analysis plan; however, according to the study protocol, changes from baseline to each pre-specified study time point were to be measured and summarized using descriptive statistics. HRQoL assessments (EQ-5D-5L and EORTC QLQ-C30) were collected during scheduled visits for all patients in cohorts A1 and F enrolled under amendment 3 and subsequent amendments (i.e., every 3 weeks ± 7 days for the first 12 weeks, in alignment with study drug administration, and every 6 weeks ± 7 days thereafter, in alignment with tumour imaging assessments) while the patient was receiving study treatment. Once a patient discontinued treatment, patient-reported outcome assessments were performed during the EOT visit, the safety follow-up visit, and the post-treatment follow-up period every 90 days (± 14 days).

For the EORTC QLQ-C30, scores and changes in scores from baseline for each domain were summarized by the number of patients with values for the mean, SD, median, range, first and third quartiles, and 95% CI. Statistical graphs were produced for each of the 15 domains to show mean scores over time, with 95% CIs. Similar graphic displays were produced for the mean change from baseline over time. In terms of missing data, if at least half of the items from a particular scale were answered, it was assumed that the missing items had values equal to the average of those items that were present. With this method of imputation, none of the single-item measures could be imputed.

Safety Outcomes

Safety and tolerability were evaluated by monitoring the frequency, duration, and severity of AEs and irAEs. In general, all by-visit summaries of safety parameters were only summarized up to and including month 6 and at the treatment discontinuation visit. AEs were organized based on the Medical Dictionary for Regularity Activities version 23.0. Only TEAEs were analyzed, but all AEs occurring during the study were listed. TEAEs were tabulated by preferred term and system organ class. Patients with the same TEAE more than once had that event counted only once for each system organ class and once for each preferred term. By-patient listings of irAEs were presented, if appropriate.

Sensitivity Analyses

No sensitivity analyses were conducted for any of the outcomes outlined in the protocol of the GARNET trial.

Subgroup Analyses

Subgroup analyses were performed for ORR, DOR, and TEAEs. The following subgroup analyses for DOR and ORR were planned a priori in the statistical analysis plan:

  • MSI status (MSI-H versus MSS versus unknown/missing)
  • histology (patients with dMMR EC only)
  • number of prior anti-cancer therapy regimens (1 versus ≥ 2)
  • prior radiation therapy (yes or no)
  • prior bevacizumab use (yes or no)
  • BOR from last platinum-containing prior anti-cancer therapy (CR or PR, SD, progressive disease, or missing)
  • progression-free interval from last platinum-containing prior anti-cancer therapy (< 6 months versus ≥ 6 months versus missing).

The following subgroups aligned with those pre-specified in the protocol for this CADTH review: histology, number of prior anti-cancer therapy regimens, prior radiation therapy, and progression-free interval from last platinum-containing prior anti-cancer therapy. Only the subgroups identified in the CADTH review protocol are reported in the Outcomes section.

Analysis Populations

The primary efficacy analysis dataset, by RECIST 1.1 per BICR, was defined as all patients in the safety analysis dataset with measurable disease at baseline (defined as the existence of at least 1 target lesion at baseline tumour assessment identified by BICR) who had the opportunity for at least 24 weeks of tumour assessment at the time of analysis (i.e., patients whose first dose of dostarlimab administration was on or before September 15, 2019).

The secondary efficacy analysis dataset, by irRECIST per investigators’ assessment, was defined as all patients in the safety analysis dataset with measurable disease at baseline (defined as the existence of at least 1 target lesion at baseline tumour assessment identified by investigator assessment) who had the opportunity for at least 24 weeks of tumour assessment at the time of analysis (i.e., patients whose first dose of dostarlimab administration was on or before September 15, 2019).

The safety analysis dataset included all patients who received any amount of study drug. In addition, all patient-reported outcome analyses were conducted on patients using the safety analysis dataset enrolled under amendment 3 or subsequent amendments.

Results

Patient Disposition

Details of patient disposition in the cohort A1 safety analysis dataset of the GARNET trial are summarized in Table 10. A total of 479 patients were screened and, of those, 129 were enrolled into cohort A1. In cohort A1, there were 126 patients with dMMR EC and 3 with MMR-unk/MSI-H identified through NGS. All patients enrolled received dostarlimab. Of enrolled patients, 100.0%, 81.4%, and 87.6% were included in the safety, primary, and secondary efficacy analysis datasets, respectively. In IA-2, a subset of 72 patients from IA-1 were included. Refer to Appendix 4 for detailed outcomes data for this subset of patients.

At the time of IA-2, the median duration of follow-up was 16.3 months. A total of 47 (36.4%) patients had withdrawn from the study. The most common reason for study discontinuation was death, which occurred in 36 patients (27.9%); progressive disease was the main cause of death. Two patients were lost to follow-up (1.6%) and 71 (55.0%) discontinued treatment. The primary reasons for treatment discontinuation were confirmed progressive disease (38.0%) followed by AEs (10.9%). Twenty-eight (21.7%) patients continued treatment with dostarlimab beyond initial confirmation of progressive disease. Thirty-six patients (27.9%) died during the study, 31 (24.0%) due to progressive disease and 5 (3.9%) due to an AE.

Table 10. Patient Disposition in Cohort A1 of the GARNET Trial (IA-2, Safety Analysis Dataset).

Table 10

Patient Disposition in Cohort A1 of the GARNET Trial (IA-2, Safety Analysis Dataset).

Protocol Deviations

Protocol deviations in cohort A1 of the GARNET trial are summarized in Table 11. There were 168 important protocol deviations in 75 (58.1%) patients in the safety analysis dataset. Most of these deviations were related to study visits or procedures (n = 61; 42.1%), There were 17 significant protocol deviations in 11 (8.5%) patients, the majority of which were related to eligibility criteria (n = 8; 6.2%). The sponsor noted that “none of these significant protocol deviations were considered to affect the patients’ safety or well-being or the overall integrity of the study.”

Table 11. Summary of Protocol Deviations in Cohort A1 of the GARNET Trial (IA-2).

Table 11

Summary of Protocol Deviations in Cohort A1 of the GARNET Trial (IA-2).

Exposure to Study Treatments

Duration and Dose Intensity

Data on exposure to dostarlimab in cohort A1 at IA-2 are summarized in Table 12. Data were available for 129 patients with dMMR or MMR-unk/MSI-H. The median DoT with dostarlimab was 25.6 weeks. The maximum DoT with dostarlimab was 138.9 weeks. Approximately half of patients (53.5%) received dostarlimab treatment for up to 19 to 24 weeks. The median relative dose intensity was 100% throughout the study treatment period because dose reductions were not permitted. Twenty-eight patients continued study treatment despite initial progressive disease.

Treatment Compliance

Treatment compliance was based on number of infusions. At any time during the study, less than 6% of patients with dMMR EC had a missed infusion, and less than 20% had an infusion delay. From cycle 1 to cycle 4, there were 3 missed infusions (2.4%) and 11 infusion delays (8.7%) reported in patients with dMMR EC. From cycle 5 through the EOT, 7 missed infusions (5.6%) and 25 infusion delays (19.8%) were reported in patients with dMMR EC. One infusion interruption (0.8%) was reported between cycle 5 and EOT.

Table 12. Patients With dMMR or MSI-H EC on Treatment by Week Intervals in Cohort A1 of the GARNET Trial (IA-2, Safety Analysis Dataset).

Table 12

Patients With dMMR or MSI-H EC on Treatment by Week Intervals in Cohort A1 of the GARNET Trial (IA-2, Safety Analysis Dataset).

Concomitant Medication

Nearly all patients in cohort A1 reported taking concomitant medications (99.2% of dMMR EC patients). The most common types of concomitant medications (used by ≥ 40% of patients) were in the Anatomical Therapeutic Chemical (ATC) classes of the following:

  • other analgesics and antipyretics (76.2%)
  • opioids (58.7%)
  • antithrombotic drugs (49.2%)
  • drugs for peptic ulcer and gastro-esophageal reflux disease (47.6%)
  • anti-inflammatory and antirheumatic products, nonsteroids (42.1%)
  • drugs for constipation (41.3%).
Subsequent Treatments

Thirty-three patients received subsequent anti-cancer therapy after treatment with dostarlimab. Of these patients, 27 had progressive disease. These therapies, from most to least frequent, included the following:

  • radiotherapy (7.8%)
  • single-drug chemotherapy, such as doxorubicin (4.7%), carboplatin (4%), paclitaxel (4%), and pegylated liposomal doxorubicin (4%)
  • immunotherapy, such as pembrolizumab (4.0%) and bevacizumab (2.4%)
  • hormone therapy, such as letrozole (3.1%), megestrol acetate (0.8%) medroxyprogesterone (0.8%), tamoxifen (0.8%), temsirolimus (0.8%)
  • combination chemotherapy, such as paclitaxel plus carboplatin (2.4%) and carboplatin plus gemcitabine (1.6%)
  • surgery (0.8%).

Outcomes

Only outcomes and analyses of the subgroups identified in the review protocol are reported below. Data tables for the subset of patients from IA-1 included in IA-2 (n = 72) can be found in Appendix 4. In addition, data tables for all immune-related outcomes (except irPFS) are available in Appendix 4. As part of the sponsor’s feedback on this CADTH reimbursement review report, the sponsor provided CADTH with an updated analysis (data cut-off of November 1, 2021) for certain baseline characteristics, efficacy, and safety outcomes in the GARNET trial. The results of the updated analysis were, overall, consistent with those reported in the previous analyses of data from the March 1, 2020, cut-off date. The additional results from the November 1, 2021, data cut-off date are available in Appendix 5.

Overall Survival

The results for OS from cohort A1 of the GARNET trial are summarized in Table 13. The median OS was not reached at the time of IA-2. The median follow-up time was 16.3 months. In total, there were 35 deaths (33.3%), and 70 patients (66.7%) were censored. The probability of patients in cohort A1 surviving to 6, 9, and 12 months was 80.9% (95% CI, 71.7% to 87.4%), 75.1% (95% CI, 65.2% to 86.6%), and 68.9% (95% CI, 58.3% to 77.4%), respectively.

Table 13. KM Analysis of OS for Patients With dMMR or MSI-H EC in Cohort A1 of the GARNET Trial (IA-2, Primary Efficacy Analysis Dataset).

Table 13

KM Analysis of OS for Patients With dMMR or MSI-H EC in Cohort A1 of the GARNET Trial (IA-2, Primary Efficacy Analysis Dataset).

Kaplan-Meier estimates of OS at IA-2 in patients with dMMR or MSI-H EC for the Secondary Efficacy Analysis Dataset. The total number of at-risk patients in the dMMR or MMR-unk/MSI-H EC at 0, 4, 8, 12, 16, 20, 24, 28, 32, and 36 months was 87, 68, 46, 37, 20, 13, 5, 0, 0, respectively.

Figure 3

KM Plot of OS for Patients With dMMR or MSI-H EC per RECIST 1.1 (IA-2, Secondary Efficacy Analysis Dataset).

Disease Control Rate and Immune-Related Disease Control Rate

The DCR results (based on investigator assessment) of the GARNET trial for cohort A1 are summarized in Table 14. The DCR was 57.1%, with 11 patients (10.5%) having a BOR of CR, 36 patients (34.3%) having a BOR of PR, and 13 patients (12.4%) having a BOR of stable disease. Forty-two (89.4%) patients at the time of IA-2 had an ongoing response.

irDCR by investigator assessment per irRECIST 1.1 (n = 113) was 63.7% for patients with dMMR or MSI-H EC, which was slightly higher than the DCR. Eight patients (7.1%) had an irBOR of irCR, 44 patients (38.9%) had an irBOR of irPR, and 20 patients (17.7%) had an irBOR of irSD. Forty-three (82.7%) patients at the time of IA-2 had an ongoing response. Detailed irDCR results are available in Appendix 4.

Table 14. Tumour Response in Patients With dMMR or MSI-H EC in Cohort A1 of the GARNET Trial per RECIST 1.1 Assessed by BICR (IA-2, Primary Efficacy Analysis Dataset).

Table 14

Tumour Response in Patients With dMMR or MSI-H EC in Cohort A1 of the GARNET Trial per RECIST 1.1 Assessed by BICR (IA-2, Primary Efficacy Analysis Dataset).

Health-Related Quality of Life
EORTC QLQ-C30

EORTC QLQ-C30 data were available for 94 of the 129 patients in cohort A1 of the GARNET trial at the time of IA-2. Completion rates for the EORTC QLQ-C30 instrument declined over time. The completion rate at baseline was 100% and 58.5% by cycle 7 (n = 55). The mean scores and mean changes from baseline at each assessment point for EORTC QLQ-C30 (global health status or quality-of-life scale) are summarized in Table 15 and Figure 4. A definition for what constituted a clinically meaningful change from baseline in the study population was not provided. Overall, summary scores increased or remained stable from baseline to cycle 7 among patients who completed questionnaires.

Table 15. Summary of EORTC QLQ-C30 Results — Global Health Status and QoL Function From Baseline to Cycle 7 (IA-2, Safety Analysis Dataset).

Table 15

Summary of EORTC QLQ-C30 Results — Global Health Status and QoL Function From Baseline to Cycle 7 (IA-2, Safety Analysis Dataset).

A summary of the disease-related symptom subscales can be found in Figure 5. Patients who received dostarlimab and completed the questionnaire reported decreased symptoms or remained stable over time in key disease-related symptom subscales, including pain and fatigue.

Mean change in symptomatic AE response from baseline over time is illustrated in Figure 6. Most patients who experienced symptomatic AEs, including nausea, vomiting, constipation, diarrhea, or tiredness, remained stable or had improvement from baseline in these symptoms over the treatment course. Less than 25% of patients reported single-category worsening in these AE symptoms, and less than 6% reported 2- or 3-category worsening.

Graph of the mean change in global health status from cycle 2 to cycle 7 at IA-2 in the safety analysis dataset.

Figure 4

Mean Change in Global Health Status and QoL From Baseline (IA-2, Safety Analysis Dataset).

Three graphs of the mean change in pain, fatigue, and physical functioning from cycle 2 to cycle 7 at IA-2 in the safety analysis dataset.

Figure 5

Pain, Fatigue, and Physical Functioning Mean Change From Baseline (IA-2, Safety Analysis Dataset).

Five graphs showing the mean change in response for symptomatic AEs (i.e., nausea, vomiting, constipation, diarrhea, and tiredness) from cycle 2 to cycle 7 at IA-2 in the safety analysis dataset.

Figure 6

Symptomatic AE Change in Response From Baseline (IA-2, Safety Analysis Dataset).

EQ-5D-5L

EQ-5D-5L data were available for 89 of 129 patients in cohort A1 at the time of IA-2. Summary data for the descriptive system of the EQ-5D-5L were not reported for any of the 5 dimensions. Overall, completion rates for the EQ-5D-5L and corresponding EQ-VAS instrument declined over time; there were 89 patients who completed questionnaires at baseline and 5 or fewer patients at each post-treatment visit.

EQ-VAS

At baseline, the mean EQ-VAS score was 69.3 (SD = 19.2) (refer to Figure 7). The following changes were observed in the EQ-VAS scores over time:

  • At week 12, the mean score was 77.1 (SD = 18.0) and mean change from baseline was 5.0 (SD = 12.6).
  • At week 18, mean score was 77.4 (SD = 17.4) and mean change from baseline was 4.0 (SD = 15.2).
  • At week 42, the change from baseline was 4.0 (SD = 16.2).

Greater change in scores was observed after EOT; however, the number of patients at each post-treatment visit was markedly low (< 5 patients).

Graph showing the adjusted mean change in EQ-VAS from baseline at IA-2 in the safety analysis dataset.

Figure 7

Adjusted Mean Change From Baseline in EQ-VAS (IA-2, Safety Analysis Dataset).

Progression-Free Survival and Immune-Related Progression-Free Survival

The results for PFS in cohort A1 of the GARNET trial, based on BICR, are summarized in Table 16. At IA-2, 57 (54.3%) patients had a PFS event and median PFS was 5.5 (95% CI, 3.2 to NR) months. KM estimates of PFS by RECIST 1.1 were 48.6% (95% CI, 38.6% to 56.8%) at month 6 and 47.5% (95% CI, 37.4% to 56.8%) at month 9 and at month 12. Refer to Figure 8 for the PFS KM curve.

In terms of irPFS, 57.3% of patients had an irPFS event at the time of IA-2. The median irPFS time was 10.3 months (95% CI 5.2 to 18.0). Based on investigator assessment, the probability of having irPFS based on KM estimates were 54.7% at 6 months, 50.7% at 9 months, and 47% at 12 months for patients with dMMR or MSI-H EC (95% CI not reported). KM estimates for irPFS were higher than that of PFS by BICR. Refer to Figure 13 in Appendix 4 or the irPFS KM curve.

Table 16. KM Analysis of PFS in Patients With dMMR or MSI-H EC in Cohort A1 of the GARNET Trial per RECIST 1.1 Assessed by BICR (IA-2, Primary Efficacy Analysis Dataset).

Table 16

KM Analysis of PFS in Patients With dMMR or MSI-H EC in Cohort A1 of the GARNET Trial per RECIST 1.1 Assessed by BICR (IA-2, Primary Efficacy Analysis Dataset).

Kaplan-Meier estimates of PFS at IA-2 in patients with dMMR or MSI-H EC for the Secondary Efficacy Analysis Dataset. The total number of at-risk patients in the dMMR or MMR-unk/MSI-H EC at 0, 4, 8, 12, 16, 20, 24, 28, 32, and 36 months was 105, 88, 57, 46, 43, 34, 30, 24, 24, 16, 12, 12, 7, 5, 2, 2, 0, 0, 0, respectively.

Figure 8

KM Plot for PFS in Patients With dMMR or MSI-H EC per RECIST 1.1 Assessed by BICR (IA-2, Primary Efficacy Analysis Dataset).

Objective Response Rate and Immune-Related Objective Response Rate

The results for ORR (based on BICR) for cohort A1 of the GARNET trial are summarized in Table 12. The ORR results were generally consistent with the results for irORR.

At the time of IA-2, the ORR, per RECIST 1.1, was 44.8% (95% CI, 35.0% to 54.8%); 47 of 105 patients achieved either a CR (n = 11) or PR (n = 36) (Table 14). The median follow-up time was 16.3 months. The ORR results were generally consistent with the results of irORR; the irORR, per irRECIST 1.1, was 46% (95% CI, 36.6% to 55.6%), with 82.7% of responders having an ongoing immune-related response at the time of IA-2.

The ORR results for pre-specified patient subgroups of interest to the CADTH review are summarized in Table 17. The ORR was 49.2% in patients with 1 line of prior anti-cancer therapy, compared with 36.8% in patients who received 2 or more lines of prior anti-cancer therapy. The ORR was 46.6% in patients with prior radiation, compared with 40.0% in patients who had not received prior radiation. The ORR was 46.0% in patients with a progression-free interval of at least 6 months, compared with 39.5% in patients with a progression-free interval of less than 6 months. The ORR was 56.3% in patients with type II endometrioid carcinoma and 40.0% in patients with type I endometrioid carcinoma. Note that 95% CIs were not provided for any of the ORR subgroup analyses.

Table 17. Subgroup Analysis Results for ORR in Cohort A1 of the GARNET Trial (IA-2, Primary Efficacy Analysis Dataset).

Table 17

Subgroup Analysis Results for ORR in Cohort A1 of the GARNET Trial (IA-2, Primary Efficacy Analysis Dataset).

The sponsor noted that there were too few patients of each endometrioid carcinoma type II subtype to make a meaningful comparison based on histologic subtype.

Duration of Response and Immune-Related Duration of Response

The DOR results (based on BICR) for cohort A1 of the GARNET trial are summarized in Table 18.

Of the 47 patients who achieved an objective response, 42 (89.4%) had an ongoing response at the time of the IA-2 data cut-off. The median duration of follow-up was 16.3 months; however, the median DOR was not reached among responders. The DOR in these patients ranged from 2.63 to 28.09 months as at the time of the IA-2data cut-off, with 37 patients (78.7% of responders) achieving a DOR of at least 6 months. Based on these results, the probability of responders maintaining a confirmed objective response, per RECIST 1.1, was estimated to be 97.9% (95% CI, 85.8% to 99.7%), 90.9% (95% CI, 73.7% to 97.1%), and 80.1% (95% CI, 56.8% to 91.7%) at months 6, 12, and 18, respectively.

The sponsor noted that the small sample size of responders hampered the ability to estimate the DOR of patient subgroups, so these data were not reported.

The results of investigator-assessed irDOR were generally consistent with DOR assessed by BICR (refer to Appendix 4 for further details). At the time of the IA-2, 43 patients had an ongoing immune-related response out of the 52 who achieved an irCR or irPR. The median irDOR was not reached among responders after a median follow-up of 16.5 months. Approximately 76.9% of responding patients achieved an irDOR of at least 6 months. Based on KM estimates, the probability of maintaining a response, per irRECIST, at 6 months was 96.1% (95% CI, 85.2 to 99.0) and at 12 months and at 18 months was 79.2% (95% CI, 62.1% to 89.2%).

Table 18. KM Analysis of DOR for Patients With dMMR or MSI-H EC in Cohort A1 of the GARNET Trial per RECIST 1.1 Assessed by BICR (IA-2, Primary Efficacy Analysis Dataset of Patients With Objective Response).

Table 18

KM Analysis of DOR for Patients With dMMR or MSI-H EC in Cohort A1 of the GARNET Trial per RECIST 1.1 Assessed by BICR (IA-2, Primary Efficacy Analysis Dataset of Patients With Objective Response).

Harms

Only those harms identified in the CADTH review protocol are reported in this section. A summary of harms data is provided in Table 19.

Adverse Events

The most frequently reported TEAEs (≥ 15%) in patients with dMMR or MSI-H EC were nausea, diarrhea, anemia, fatigue, asthenia, constipation, vomiting, abdominal pain, cough, arthralgia, urinary tract infection, and back pain. With the exception of anemia, these TEAEs were reported to be mild or moderate in severity in most patients.

Grade 3 or higher TEAEs occurred in 48.1% of patients in cohort A1. The most commonly reported grade 3 or higher TEAEs were anemia (14.7%), abdominal pain (5.4%), and hyponatremia (3.9%). Grade 4 TEAEs were reported for hyponatremia, pulmonary embolism, pneumonia, and sepsis. Sepsis was reported as a grade 4 TEAE in 3 patients with dMMR EC and 1 patient with MMR-unk/MSI-H. Other grade 3 or higher TEAEs included acute kidney injury (3.1%) and back pain (3.1%).

Serious Adverse Events

The percentage of patients that experienced a serious TEAE was 34.1% in cohort A1. The most common serious TEAEs were abdominal pain, acute kidney injury, and sepsis, each occurring in 3.1% of patients, and pulmonary embolism, pyrexia, and urinary tract infection, each occurring in 2.3% of patients.

Withdrawals Due to AEs

None of the patients in cohort A1 who withdrew from the GARNET trial had an AE as a primary reason. AEs that led to study treatment discontinuation occurred in 11.6% of patients, whereas AEs that led to study treatment interruption occurred in 24.0% of patients. The most common AEs leading to study treatment interruption were anemia (3.1%) and diarrhea (2.3%).

Mortality

TEAEs leading to death occurred relatively rarely in cohort A1 during the treatment period (n = 1; 0.8%) and included aspiration. TEAEs leading to death during the 90-day safety follow-up occurred in 4 patients (3.1%), and included pleural effusion, pneumonia, sepsis, and shock. None of the TEAEs leading to death were considered by the investigators to be treatment-related. No TEAEs were the primary cause of death during the long-term follow-up period.

Notable Harms

Notable harms as specified in the CADTH review protocol included immune-related toxicity.

The incidence of irAEs was 34.9% in cohort A1. The most frequently reported irAEs (≥ 5%) were diarrhea and hypothyroidism. A total of 7.9% of patients had a serious irAE, 12.7% had an irAE that was grade 3 or higher, and 4.8% of patients with dMMR EC had an irAE that led to study treatment discontinuation. Most of the irAEs were considered by the investigators to be related to study treatment.

Table 19. Summary of Harms in Cohort A1 of the GARNET Trial.

Table 19

Summary of Harms in Cohort A1 of the GARNET Trial.

Critical Appraisal

Internal Validity

The main limitation of the GARNET trial stems from the single-arm trial design and the lack of a comparator group. Such a design makes it challenging to interpret the efficacy and safety events attributable to dostarlimab, because all patients in cohort A1 received the same treatment. The lack of comparison with an active comparator or with standard of care or placebo precludes the ability to assess the relative therapeutic benefit or safety of dostarlimab. Formal statistical significance and hypothesis-testing were not performed, except for ORR. The risk of selection bias cannot be ruled out, given the lack of transparency regarding the selection and timing of the enrolment of patients in the trial. In the absence of such information, the risk of bias is unclear. GARNET is an open-label trial, and the study investigators and patients were aware of their treatment status, which increases the risk of detection and performance bias; this has the potential to influence results and outcomes in favour of dostarlimab if the assessor (investigator or patient) believes the study drug is likely to provide a benefit. Furthermore, if study personnel and patients knew that the treatment was dostarlimab (which is known to cause irAEs and other neurotoxicity), this also could have influenced the reporting of harms outcomes. To mitigate the impact of such bias, the investigators used a BICR to evaluate responses using standardized criteria for certain efficacy outcomes, such as ORR, DOR, PFS, and DCR. Bias is therefore less a concern for these efficacy end points and OS, and more of a concern for subjective end points such as HRQoL and safety. Overall, the magnitude and direction of this bias remain unclear.

Median OS was not reached at the time of IA-2, so the survival data from the trial were immature. Because results were based on an interim analysis, the treatment benefit may be overestimated and harms may be underestimated. In addition, the potential for confounding cannot be ruled out, given the lack of adjustment in the analysis for known confounders and treatment-effect modifiers that would typically be accounted for in an RCT. Interpretation of time-to-event end points such as OS or PFS is limited in single-arm studies. The clinical experts agreed that in the absence of robust comparative data on PFS and OS, no firm conclusions could be drawn on how dostarlimab compares with other relevant treatment options.

The open-label, single-arm design also limits the ability to interpret HRQoL data from the GARNET trial. Loss of patients completing HRQoL questionnaires over the course of the study led to small sample sizes at many assessment time points (in some cases < 5 patients) and resulted in imprecise measurement, with wide and overlapping CIs. Statistical analyses of changes in scale scores over time were not conducted, and a definition of what was considered a clinically meaningful response or minimally important difference was not reported. As well, summary data were not reported for the EQ-5D-5L descriptive system. For these reasons, no conclusions should be drawn from the HRQoL data. This is considered a significant limitation of the GARNET trial, because HRQoL was identified as an important outcome by the patient and clinician groups providing input for this review.

External Validity

Overall, the clinical experts consulted by CADTH agreed that the inclusion and exclusion criteria, baseline patient characteristics, concomitant medications, and prohibited medications in cohort A1 of the GARNET trial were reflective of patients they see in clinical practice for the indication under review. There were no barriers to the identification of patients who would most benefit from the treatment, given that testing for MMR and MSI status is standard practice in Canada. The population enrolled in the trial was consistent with the population expected to be treated in Canadian clinical practice, but the clinical experts noted that patients in the GARNET trial were slightly younger than those seen in clinical practice. The clinical experts also noted that patients with certain comorbidities (e.g., obesity) and of certain racial groups (e.g., Black) may have poorer prognosis; however, the findings of this study may not necessarily be generalizable to these subgroups, given the small sample size and lower level of representation of ethnic groups in the study (n = 2). Furthermore, the clinical experts indicated that no different treatment effect would be expected based on different disease-management practices in participating countries. In the opinion of the clinical experts, as long as patients have dMMR or MSI-H tumour status, dostarlimab would be appropriate to administer after any of the prior therapies received by patients in the trial. The majority of patients (88%) in cohort A1 of the GARNET trial had received 2 or fewer prior lines of systemic therapy before trial enrolment, and 11% of patients had received 3 or more lines of prior systemic therapy. However, the clinical experts noted that experience from clinical practice suggests that dostarlimab may have a lower magnitude of treatment benefit in patients with more prior lines of systemic therapy. This aligns with the results of the ORR subgroup analysis, in which the ORR was higher in patients with 1 line of prior anti-cancer therapy than in patients with 2 or more lines of prior anti-cancer therapy (49.2% versus 36.8% [95% CI, NR to NR]). However, these results are exploratory, and conclusions should be interpreted with caution. The clinical experts and clinician groups agreed that patients should not have previously been treated with immunotherapies. In cohort A1, 3 patients could not have their MMR tumour status determined, but their tumours were classified as MSI-H. The clinical experts noted that these 2 groups are generally thought to be synonymous, and data could be pooled together, especially given the small number of patients with MSI-H status. Concomitant medications received by patients in the trial appeared to reflect the medications that patients would receive in Canadian clinical practice, according to the clinical experts. It is also acknowledged that mature OS data will be confounded by the use of subsequent anti-cancer therapy received by some patients after progressive disease.

According to the clinical experts consulted by CADTH, OS, PFS, and HRQoL are clinically meaningful end points for patients with dMMR or MSI-H recurrent or advanced EC that has progressed on or after treatment with a platinum-containing regimen. Tumour response outcomes are also important in this patient population because of the accompanying delay in the worsening of symptoms and the slower decline in ECOG performance status.

There were a limited number of patients in the primary efficacy dataset (N = 105). The magnitude of the treatment-effect estimates observed in a small study sample may not be replicable in a larger study sample or generalizable to the target population in real-world clinical practice. Furthermore, the subgroup analyses had no statistical comparisons and even smaller sample sizes, which limits the generalizability to a broader population. These data should be considered exploratory in nature, and no conclusions should be drawn based on the results.

Summary Indirect Evidence

Objectives and Methods for the Summary of Indirect Evidence

The objective of this section is to provide an appraisal and summary of the indirect evidence submitted by the sponsor, which included 6 reports of ITCs (3 reports of MAICs and 3 of IPTW analyses),19 which compare dostarlimab with other treatments used for patients with advanced or recurrent EC that has progressed on or after a platinum-based regimen. Results for the MAIC and IPTW analyses are presented separately.

A focused literature search for network meta-analyses dealing with EC was run in MEDLINE All (1946–) on November 3, 2021. No limits were applied to the search. The literature search identified 31 citations, none of which met the eligibility criteria. Given the lack of comparative evidence, and the phase I, single-arm nature of the GARNET trial, the sponsor-submitted ITCs were used to inform the pharmacoeconomic model. The submitted analyses were appraised and summarized.

Matching-Adjusted Indirect Comparisons

Description of MAIC Analyses

The sponsor submitted 3 MAIC reports to demonstrate the efficacy and safety of dostarlimab compared with relevant treatments for advanced or recurrent EC19:

  • Two MAIC reports that compared individual patient data (IPD) from the phase I GARNET trial with a GARNET-like real-world evidence (RWE) cohort in the UK that received current treatment paradigms (hereafter called MAIC report 1), and a selection of 5 specific treatments for advanced or recurrent EC (hereafter called MAIC report 2) from the National Cancer Registration and Analysis Service (NCRAS) database.19
  • One MAIC report that compared IPD from the phase I GARNET trial with relevant comparator data from the published literature (hereafter called MAIC report 3).19
Methods of MAIC Analyses
Objectives

In the absence of direct comparative evidence from trials, the aim of each analysis was to compare the efficacy and safety (e.g., OS, PFS, time on treatment, ORR, and/or SAEs) of dostarlimab with current UK treatment paradigms based on RWE cohorts and published literature, using the MAIC method.19

Study Selection Methods

The index trial in all cases was based on IPD from cohort A1 of part 2B of the phase I, single-arm GARNET trial (dMMR or MSI-H EC cohorts, safety population set; n = 129).19 Study selection methods to identify relevant comparators varied in the 3 sponsor-submitted MAIC reports.

For the 2 reports that used RWE as a comparator (MAIC reports 1 and 2), no systematic literature review (SLR) was conducted. Instead, the investigators created GARNET-like cohorts based on a subgroup of patients from the NCRAS in the UK, including a base-case analysis of current treatment paradigms, an analysis of 5 treatment-specific cohorts, and a sensitivity analysis of patients with an ECOG Performance Status of 0 or 1 at index. Notably, information on dMMR or MSI-H status (an inclusion criterion in GARNET and the indication under review) was not available in the RWE cohorts. In MAIC report 3, a SLR of clinical and observational studies was conducted in accordance with the highest standard for evidence synthesis (date last searched, April 5, 2020).51 Although there were no limitations to study eligibility by risk of bias, the authors noted that the included studies were generally at low risk of bias per the NICE tool,52 and bias was either high or unclear per the CASP tool.19,53

The selection characteristics used to develop the GARNET-like cohorts and the population, intervention, comparison, outcomes, and study (PICOS) criteria used to identify published literature are summarized in Table 20.

Table 20. Selection Criteria for GARNET-Like Subgroups and for Relevant Comparator Studies for Sponsor-Submitted MAICs.

Table 20

Selection Criteria for GARNET-Like Subgroups and for Relevant Comparator Studies for Sponsor-Submitted MAICs.

The outcomes evaluated in the MAICs are summarized in Table 21. OS was the pre-specified primary outcome in all MAICs. In MAIC report 3, PFS was also included as a pre-specified primary outcome. PFS and TTD were the pre-specified secondary outcomes of MAIC reports 1 and 2. These were summarized descriptively, as the measurement definitions for PFS and the time period evaluations associated with the RWE cohort were too dissimilar to those in GARNET. As PFS was not recorded in the NCRAS database, TTNT and TTD were used as proxies. Pre-specified secondary end points in MAIC report 3 included ORR, DOR, HRQoL, and SAEs; however, no definitions were provided for these outcomes in the report.19

Table 21. End Point Definitions in Included Studies.

Table 21

End Point Definitions in Included Studies.

MAIC Analysis Methods

Given that the GARNET trial was a single-arm trial, unanchored MAICs were necessary to conduct the ITCs because the single-arm nature precluded the use of anchored ITC methods. A summary of the analysis methods in each MAIC is provided in Table 23.

GARNET Versus NCRAS RWE Cohort MAIC Methods (MAIC Reports 1 and 2)

Analysis methods for the MAICs conducted using RWE data (MAIC reports 1 and 2) were identical, only differing in the RWE cohorts used. The GARNET trial was a phase I, open-label, dose-escalation study, whereas the RWE study was a descriptive, noninterventional study of patient-level data available through the NCRAS to describe characteristics, treatments, and outcomes of patients diagnosed with advanced or recurrent EC. The dataset for analysis consisted of IPD for all patients in the GARNET safety cohort (n = 129), and aggregated summary information from the initial RWE cohort of incident patients at second-line treatment (n = 999; n = 501 for patients with an ECOG Performance Status of 0 or 1 at index). Treatment-specific RWE cohorts included patients who received paclitaxel monotherapy (n = 116), carboplatin plus paclitaxel (n = 279), carboplatin plus liposomal doxorubicin (n = 141), liposomal doxorubicin monotherapy (n = 130), and carboplatin monotherapy (n = 93).19

Key differences were noted by the sponsor between the GARNET trial and the NCRAS RWE cohort’s inclusion criteria. One of the main differences between GARNET and the RWE cohort was the MMR or MSI status of patients. All patients in GARNET had dMMR or MSI-H EC, whereas this information was not available in the RWE cohort, so patients were included regardless of MMR or MSI status. Another key difference between GARNET and the RWE cohort’s inclusion criteria was that the RWE cohort used platinum doublet therapy as the key milestone regarding lines of therapy. The RWE cohort did not analyze the lines of therapy before the diagnosis of advanced or recurrent EC. The first-line therapy received for advanced or recurrent diagnosis was platinum doublet therapy for nearly all (99.3%) patients. Thus, the incident population at second-line therapy gave rise to the base case sample size (n = 999). Last, although all patients in GARNET had an ECOG Performance Status of 0 or 1, the inclusion criteria on ECOG Performance Status were partially met in the RWE cohort. In GARNET, ECOG Performance Status was ascertained at the index date of the trial (i.e., the start of second-line therapy), whereas in the RWE cohort, ECOG Performance Status was recorded at diagnosis. Also, there were many patients with ECOG Performance Status documented as “not recorded” in the RWE cohort. A sensitivity analysis conducted with the RWE cohort was restricted to patients with a known ECOG Performance Status of 0 or 1. This was referred to as the RWE cohort (ECOG ≤ 1).19

A total of 8 analyses (4 base-case and 4 scenario analyses) were conducted in the initial RWE cohort of MAIC report 1. Hazard ratios (HRs), 95% CIs, and corresponding P values were calculated only for the OS outcome (comparisons were descriptive for other outcomes), as follows19:

  • OS in GARNET (N = 129) versus OS in RWE cohort (base case, n = 999; ECOG ≤ 1, n = 501)
  • PFS in GARNET (N = 129) versus TTNT in RWE cohort (base case, n = 999; ECOG ≤ 1, n = 501)
  • PFS in GARNET (N = 129) versus TTD in RWE cohort (base case, n = 999; ECOG ≤ 1, n = 501)
  • TTD in GARNET (N = 129) versus TTD in RWE cohort (base case, n = 999; ECOG ≤ 1, n = 501).

A total of 4 analyses were performed for each treatment-specific RWE cohort in MAIC report 2. HRs, 95% CIs, and P values were calculated only for the OS outcome (comparisons were descriptive for other outcomes), as follows19:

  • OS in GARNET (N = 129) versus OS in treatment-specific RWE cohorts
  • PFS in GARNET (N = 129) versus TTNT in treatment-specific RWE cohorts
  • PFS in GARNET (N = 129) versus TTD in treatment-specific RWE cohorts
  • TTD in GARNET (N = 129) versus TTD in treatment-specific RWE cohorts.

A range of prognostic variables typically associated with survival in EC were identified in a targeted literature review (conducted May 2020), and validated through a panel of oncologists in Canada, Germany, and the UK, as follows19:

  • race and/or ethnicity (Black, other, unknown versus White)
  • age category (≥ 65 years versus < 65 years)
  • ECOG Performance Status at treatment initiation (1 versus 0)
  • histology at initial diagnosis (nonendometrioid, unknown versus endometrioid)
  • FIGO stage (stages III and IV versus stages I and II)
  • grade of disease at diagnosis (grades 3 and 4, unknown versus grades 1 and 2)
  • number of prior platinum-based therapies (0 or 1 versus ≥ 2)
  • prior surgery for study indication (yes versus no).

In MAIC report 1, assessment of the effect modification and prognostic value of each potential matching variable was done using a Cox proportional hazard model for each outcome (OS, PFS, and TTD). Cox regression models were fit separately for the GARNET data and the RWE data. Patient characteristics that exhibited an association at a level of significance of at least 0.1 in at least 1 of the 2 datasets were considered prognostic. P values evaluated in a multivariable model were obtained from a full model (including all variables listed above), and then backward stepwise selection, with a P ≤ 0.1 threshold, was applied. Effect-modifying status was assessed semi-empirically by comparing regression coefficients (or HRs) for the variable of interest from the 2 regression models fitted to the GARNET and the RWE dataset, where large differences between coefficients were considered to be potentially effect-modifying. The following variables were considered prognostic for each outcome19:

  • OS: ECOG, race and/or ethnicity, grade, histology, stage at diagnosis, and prior surgery
  • PFS: ECOG, and histology
  • TTNT: Stage at diagnosis, grade, and prior surgery
  • TTD: ECOG, race and/or ethnicity, grade, histology, and prior surgery.

The IPD from the GARNET trial were matched to the RWE cohort using the selected matching variables to estimate the above comparisons. A method of moments, as introduced by Signorovitch et al. (2010),54 was used to allow a propensity score logistic regression model to be estimated without IPD for the RWE cohort.19 After matching, re-weighted outcome data from the GARNET trial and RWE cohort were compared. This included calculating KM survival estimates for OS, PFS, TTD, and TTNT at given time points after index treatment initiation, as well as creating KM curves. The primary end point analysis used a Cox proportional hazard model, with weights obtained using the MAIC method, to re-estimate HRs for dostarlimab in the GARNET trial versus the RWE cohort. In the treatment-specific RWE cohort, weighted Cox proportional hazard models were fitted, and OS HRs for dostarlimab versus the 5 comparator treatments of interest were re-estimated, together with 95% CIs and P values. For use in these regression models, OS data from the RWE cohort were digitized, and pseudo-IPD were constructed from KM curves using the method described by Guyot et al. (2012).55 Log-cumulative hazards, as well as Schoenfeld residuals, were plotted for OS to examine the proportional hazards assumption (PHA) for the treatment group variable before and after matching.19

To assess the impact of each potential covariate on effective sample size (ESS) and estimated treatment effect, a “leave 1 out” approach was applied. This approach started with the calculation of ESS and a measure of treatment effect (HR for OS in the case of this study) for a full model, and then the calculation of the same 2 measures after removal of the covariate from the model; all remaining matching variables in the model were kept.19

The following matching scenarios were considered in both MAICs of the RWE cohorts19:

  • Scenario 1 included all matching variables considered most relevant by expert opinion (grade, histology [1 patient from GARNET with unknown histology was removed], number of prior platinum-based therapies in the advanced or recurrent setting [patients from GARNET with 0 or > 2 therapies were removed]).
  • Scenario 2 included all matching variables from scenario 1 except grade, because of poor reporting of grade in the RWE cohort (histology, number of prior platinum-based therapies in the advanced or recurrent setting). Scenario 2 was considered to be a sensitivity analysis of scenario 1 that was supposed to maintain a larger ESS.
  • Scenario 3 included the matching variables that were considered prognostic, based on the empirical regression analyses, with the exception of grade. Moreover, ECOG Performance Status was only used as a matching variable in the sensitivity analysis for patients with an ECOG Performance Status of 0 or 1 from the RWE cohort.

GARNET Versus Published Literature MAIC Methods (MAIC Report 3)

Three studies were eligible for the MAIC (MAIC report 3) based on the SLR. GARNET was a phase I single-arm trial of dostarlimab. McMeekin et al. (2015)56 was a global, open-label, phase III RCT comparing ixabepilone with paclitaxel or doxorubicin, Julius et al. (2013)57 was a retrospective review of medical records in the US focusing on pegylated liposomal doxorubicin, and Mazgani et al. (2008)58 was a retrospective analysis of carboplatin plus paclitaxel in patients with relapsed EC at 1 centre in Vancouver, British Columbia. The individual comparator studies were assessed for eligibility based on reporting of outcomes and appropriateness of the MAIC based on sample size and population (had to be adequately comparable to GARNET).19 Differences in eligibility criteria across studies were not discussed; however, MMR and MSI-H status was not accounted for in the comparator studies.

The specific (regression-type) modelling approach used in the analyses depended on the outcome data type. All P values presented were exploratory in nature and not conclusive. The tests performed were conducted post hoc, were not formally powered, and were not adjusted for multiplicity, so the reported P values are not confirmatory. When available, KM curves were digitized (using GetData Graph Digitizer 2.26) and the algorithm of Guyot et al. (2012)55 was used to produce reconstructed IPD for the aggregate trial data. As the GARNET trial had less than 36 months of follow-up for OS, KM data from each digitized comparator study were restricted (censored) to 36 months or less to improve the comparability of results.19

For all studies, each KM curve was digitized separately and the reconstructed IPD were synthesized. Curves created from these reconstructed IPD were plotted against curves displaying the raw digitized survival probability time points, and visual comparisons were made against the published curves. These pseudo-IPD were considered alongside the IPD from the GARNET trial. A weighted Cox regression was then applied to the dataset, combining the reconstructed IPD with the GARNET data (no covariates other than treatment). To assess the assumption of proportional hazards in the Cox survival modelling between the treatment groups compared, a (MAIC-weighted) log-cumulative hazard plotted against log time was visually assessed for parallelism, and Schoenfeld residuals plots were produced. If the PHA was violated, an accelerated-failure-time model with a Weibull distribution was used.19

A method of moments analytical technique was used to produce individual patient-specific weights for patients from the GARNET trial to produce weighted prognostic means. Comparison of means between the aggregate trial data before and after adjustment of the GARNET trial, as well as the ESS were presented.19

In MAIC report 3, pre-specified prognostic factors were identified in the same manner as for the RWE cohort MAICs. Additionally, BMI, number of prior anti-cancer regimens, and MMR or MSI molecular profile were considered to be prognostic factors but were not included in weighting. Because of limitations of data availability, the GARNET data could only be matched on a small number of prognostic factors for McMeekin et al. (2015)56 (age, ECOG, histology, race), Julius et al. (2013)57 (age, race, number of prior chemotherapies), and Mazgani et al. (2008)58 (histology).19 The distribution of baseline characteristics by treatment group were summarized before and after matching.

To minimize heterogeneity between separate study populations before statistical adjustments, the comparator study inclusion and exclusion criteria were applied to the GARNET IPD. The definition of outcomes in the comparator studies were also compared. Disease expertise was sought to identify the most likely criteria to have been used in each study.19 The inclusion criteria of GARNET that were modified to match the comparator studies and the resulting modified sample sizes are summarized in Table 22.

Table 22. Criteria and Sample Size Used in Each MAIC Published in the Literature (MAIC Report 3).

Table 22

Criteria and Sample Size Used in Each MAIC Published in the Literature (MAIC Report 3).

No sensitivity or subgroup analyses were conducted for MAIC report 3.

Table 23. Summary of MAIC Analysis Methods.

Table 23

Summary of MAIC Analysis Methods.

Results of MAIC Analyses
Summary of Included Studies

Key design characteristics and sources of heterogeneity of the studies included in the RWE cohort MAICs and the published literature MAICs are summarized in Table 24.

GARNET Versus NCRAS RWE Cohort (MAIC Reports 1 and 2)

In total, the GARNET cohort included 129 patients and the base-case RWE cohort included 999 patients. The RWE cohort used for the sensitivity analysis (ECOG ≤ 1) included 501 patients.19

There was some variation in the baseline characteristics of patients in GARNET and MAIC reports 1 and 2 (initial and treatment-specific RWE cohorts, respectively). Age range in the study cohorts varied, with 31.2%% to 51.2% of patients younger than 65 years and with 48.8% to 68.8% of patients older than 65 years. The majority of patients were White (GARNET = 76.0%; RWE cohorts = 75.0% to 92.5%); however, the GARNET trial had a greater proportion of unknown race (15.5% versus 0.7% to 5.4%). The proportion of patients with endometrioid histology was greater in GARNET (69.8%) than in the initial RWE cohort and treatment-specific cohorts (37.6% to 44.6%). In general, the GARNET trial had a notably higher proportion of patients with an ECOG Performance Status of 0 or 1 (ECOG status for many patients in the RWE cohorts was unknown), FIGO stage I/II (44.2% versus 15.8% to 35.4%), and grade 1/2 disease (67.4% versus 22.4% to 29.0%). The number of prior platinum-based therapies in the advanced or recurrent setting was 1 for all patients in the RWE cohort, whereas in the GARNET ITT cohort, it was 0 or 2 or more for some patients, based on the method of recording prior treatment for the RWE cohort. All patients in the GARNET trial were dMMR or MSI-H. There was no information on MMR or MSI-H status in the initial RWE cohort population.19

GARNET Versus Published Literature (MAIC Report 3)

In MAIC report 3, comparing GARNET with published literature, 23 records representing 14 trial publications and 13 unique studies were included in the SLR. Three unique studies — McMeekin et al. (2015),56 Julius et al. (2013),57 and Mazgani et al. (2008)58 — were eligible for the MAIC. It was noted that several identified studies were not deemed appropriate for a MAIC, primarily due to either a low sample size or the study population being systemically different than the GARNET trial.19

The GARNET, Julius et al. (2013),57 and Mazgani et al. (2008)58 studies were nonrandomized studies of dostarlimab, pegylated liposomal doxorubicin, and carboplatin plus paclitaxel, respectively, whereas the McMeekin et al. (2015)56 study was an open-label, RCT comparing ixabepilone with paclitaxel or doxorubicin. Only the treatment arm of paclitaxel or doxorubicin was included in the MAIC with McMeekin et al. (2015).56 The sample size of included studies ranged from 31 to 248 patients.19

An assessment of the baseline characteristics before weighting was not provided in MAIC report 3. The majority of baseline characteristics that were considered important, including age range, FIGO stage, disease grade, prior platinum-based therapy, and surgery, were not reported in the published literature. When reported, race was relatively similar between GARNET and the published literature trials. When reported, the proportion of patients with endometrioid histology and the proportion of patients with an ECOG Performance Status of 0 and 1 was greater in GARNET than in the studies included in the published literature MAIC.19

Results

GARNET Versus NCRAS Initial RWE Cohort (MAIC Report 1)

OS, PFS, and TTD in the GARNET ITT and initial RWE cohort are summarized in Table 25. For the initial RWE cohort, matching scenario 1, where studies were matched on histology, grade, and number of prior platinum-based therapies, resulted in the smallest ESS, with just 34 patients. The ESS was higher for scenario 2 and scenario 3, which did not include grade for 74 and 75 patients, respectively.19

Under all matching scenarios, dostarlimab was favoured over the current treatment paradigm for OS (scenario 1 HR = 0.52 [95% CI, 0.29 to 0.92]; scenario 2 HR = 0.35 [95% CI, 0.22 to 0.55]; scenario 3 HR = 0.31 [95% CI, 0.20 to 0.49]). Results for the sensitivity analysis of patients with an ECOG Performance Status of 0 or 1 were consistent with the base case. For PFS, 2 analyses were conducted: PFS from GARNET compared with TTNT from the RWE cohort; and PFS from GARNET compared with TTD from the RWE cohort. In general, the median PFS and PFS rate at 6, 12, and 18 months in all scenarios was higher than the comparator of current treatment paradigm, particularly for the analysis comparing GARNET PFS with RWE TTD. The median TTD and TTD rates at 6, 12, and 18 months were higher in all scenarios for dostarlimab than in the RWE cohort.19

The analysis of the leave-1-out approach to assess the impact of each potential covariate on ESS and estimated treatment effect is summarized for OS in Table 26. Omission of disease grade had the largest impact on ESS and estimated treatment effect, as a substantial proportion of patients in the RWE cohort had unknown grade.19

Table 24. Key Study and Baseline Characteristics in the GARNET and Comparator Studies.

Table 24

Key Study and Baseline Characteristics in the GARNET and Comparator Studies.

Table 25. Survival Outcomes Before and After Matching — GARNET Versus Initial RWE Cohort (MAIC Report 1) .

Table 25

Survival Outcomes Before and After Matching — GARNET Versus Initial RWE Cohort (MAIC Report 1) .

Table 26. Summary of Leave-1-Out Approach — Initial RWE Cohort (Base-Case and Scenario Analyses, MAIC Report 1) .

Table 26

Summary of Leave-1-Out Approach — Initial RWE Cohort (Base-Case and Scenario Analyses, MAIC Report 1) .

GARNET Versus NCRAS Treatment-Specific Cohorts (MAIC Report 2)

For the treatment-specific MAICs, Table 27 summarizes the OS for dostarlimab compared with the 5 specific treatments in the RWE cohort. All results for scenario analyses using unweighted GARNET data were consistent with the primary analyses for OS. For paclitaxel monotherapy, the ESS in scenarios 1, 2, and 3 was 30, 72, and 63, respectively. Under all matching scenarios, the median OS (and HRs) favoured dostarlimab over paclitaxel monotherapy (scenario 1: HR = 0.36; 95% CI, 0.19 to 0.65; scenario 2: HR = 0.24; 95% CI, 0.15 to 0.40; scenario 3: HR = 0.18; 95% CI, 0.11 to 0.30), indicating that patients taking dostarlimab had a lower risk of death than patients receiving paclitaxel. The OS rates at 6, 12, and 18 months were greater for dostarlimab than for paclitaxel in the RWE cohort.19

For carboplatin plus paclitaxel, the ESS in scenarios 1, 2, and 3 was 36, 74, and 69, respectively. Dostarlimab was favoured over carboplatin plus paclitaxel only in scenarios 2 and 3 (scenario 2: HR = 0.48; 95% CI, 0.29 to 0.78; scenario 3: HR = 0.42; 95% CI, 0.25 to 0.68), but not scenario 1 (HR = 0.70; 95% CI, 0.39 to 1.28). The OS rates at 6, 12, and 18 months were greater for dostarlimab than for carboplatin plus paclitaxel in the RWE cohort.19

For carboplatin plus liposomal doxorubicin, the ESS in scenarios 1, 2, and 3 was 26, 69, and 74, respectively. Dostarlimab was favoured over carboplatin plus liposomal doxorubicin only in scenarios 2 and 3 (scenario 2: HR = 0.45; 95% CI, 0.27 to 0.76; scenario 3: HR = 0.40; 95% CI, 0.24 to 0.67), but not scenario 1 (HR = 0.74; 95% CI, 0.39 to 1.41). After matching, the 6-, 12-, and 18-month OS rates were greater for dostarlimab.19

For liposomal doxorubicin monotherapy, the ESS in scenarios 1, 2, and 3 was 37, 78, and 76, respectively. Under all matching scenarios, the median OS (and HRs) favoured dostarlimab over liposomal doxorubicin monotherapy (scenario 1: HR = 0.20; 95% CI, 0.09 to 0.44; scenario 2: HR = 0.17; 95% CI, 0.10 to 0.29; scenario 3: HR = 0.16; 95% CI, 0.09 to 0.27), indicating that patients taking dostarlimab had a lower risk of death than patients taking liposomal doxorubicin. Under all matching scenarios, the OS rates at 6 and 12 months were greater in the GARNET ITT cohort than in the RWE cohort.19

For carboplatin monotherapy, the ESS in scenarios 1, 2, and 3 was 23, 67, and 69, respectively. Dostarlimab was favoured over carboplatin in all matching scenarios (scenario 1: HR = 0.53; 95% CI, 0.29 to 0.98; scenario 2: HR = 0.32; 95% CI, 0.19 to 0.55; scenario 3: HR = 0.28; 95% CI, 0.16 to 0.48), indicating that patients taking dostarlimab had a lower risk of death than patients taking carboplatin monotherapy. Under all matching scenarios, the OS rates at 6, 12, and 18 months were greater in the GARNET ITT cohort compared with in the RWE cohort.19

Table 27. OS Before and After Matching — GARNET Versus Treatment-Specific RWE Cohort (MAIC Report 2).

Table 27

OS Before and After Matching — GARNET Versus Treatment-Specific RWE Cohort (MAIC Report 2).

Results for PFS for all comparators are summarized in Table 28. When compared with the RWE cohort TTNT and TTD, the median PFS was longer for dostarlimab than for all relevant comparators in all scenarios except scenario 1 of the TTNT comparison for paclitaxel monotherapy, carboplatin plus paclitaxel, carboplatin plus liposomal doxorubicin, and carboplatin monotherapy. For the TTD comparison, dostarlimab was favoured over all comparators in all scenarios except carboplatin monotherapy in scenario 1. The PFS rate for dostarlimab was greater in all scenarios and at all time points for the TTD comparison for all relevant comparators. In contrast with TTNT from the RWE cohort, the PFS rate for dostarlimab was greater than both paclitaxel and liposomal doxorubicin monotherapy at all time points and in all scenarios. The PFS rate was not greater at the 6-month assessment in any scenario for carboplatin plus paclitaxel, carboplatin plus liposomal doxorubicin, or carboplatin monotherapy. Otherwise, dostarlimab had a greater PFS rate at the 12- and 18-month time points in all scenarios.19

Table 28. PFS Before and After Matching — GARNET Versus Treatment-Specific RWE Cohort (MAIC Report 2).

Table 28

PFS Before and After Matching — GARNET Versus Treatment-Specific RWE Cohort (MAIC Report 2).

For the outcome of TTD (Table 29), the median TTD and TTD rates were longer for dostarlimab than for all relevant comparators in the RWE cohort at all time points and across all 3 matching scenarios.19

Table 29. TTD Before and After Matching — GARNET Versus Treatment-Specific RWE Cohort (MAIC Report 2).

Table 29

TTD Before and After Matching — GARNET Versus Treatment-Specific RWE Cohort (MAIC Report 2).

GARNET Versus Published Literature (MAIC Report 3)

Table 30 presents the ESS and weighted baseline characteristics for the MAICs versus published literature sources. A full list of baseline characteristics in GARNET after matching is summarized in Table 52. Seven patients were removed from GARNET who did not meet inclusion or exclusion criteria for McMeekin et al. (2015),56 resulting in a MAIC base of 122 patients. These 122 patients from GARNET were then weighted for each prognostic variable available, resulting in an ESS of 87.274. Because of a lack of information on inclusion and exclusion criteria in Julius et al. (2013),57 no patients were removed from the GARNET trial before weighting. After matching on available prognostic variables, the resulting ESS compared with Julius et al. (2013)57 was 81.992. After the removal of 39 GARNET patients who did not meet the inclusion or exclusion criteria for Mazgani et al. (2008),58 the resulting MAIC base was 90 patients, with an ESS after matching of 29.08.19

The covariate values used to derive the weights in the MAICs included age, ECOG, race, histology, and number of prior chemotherapies. Covariates not used in weighting varied across studies. Individual MAICs were conducted for each study identified in the literature.19

Table 30. Sample Size and Baseline Characteristics After Matching (MAIC Report 3).

Table 30

Sample Size and Baseline Characteristics After Matching (MAIC Report 3).

The median OS and PFS for dostarlimab (GARNET) before and after matching compared with data from each individual study are summarized in Table 31. In all cases, the median OS was not reached before or after adjustment for the dostarlimab group. After adjustment, OS favoured dostarlimab over paclitaxel and over doxorubicin using data from the McMeekin et al. (2015)56 study (HR = 0.407; 95% CI, 0.252 to 0.657) and over pegylated liposomal doxorubicin using data from the Julius et al. (2013)57 study (HR = 0.287; 95% CI, 0.170 to 0.486). After adjustment, there was no difference in OS for dostarlimab compared with carboplatin plus paclitaxel using data from the Mazgani et al. (2008)58 study (HR = 0.559; 95% CI, 0.256 to 1.220). Data for PFS were only available from the Mazgani et al. (2008)58 study. After adjustment, there was no difference in PFS between dostarlimab and carboplatin plus paclitaxel.19

For OS, the test of the PHA did not show a violation for the comparisons with data from the McMeekin et al. (2015),56 Julius et al. (2013),57 or Mazgani et al. (2008)58 studies (P = 0.28, 0.17, and 0.62, respectively). A Cox proportional hazards model with MAIC was fitted to the data. The HRs (0.407; 95% CI, 0.252 to 0.657 and 0.287; 95% CI, 0.170 to 0.486) indicated a difference between the 2 treatments for OS, with a 59.3%, and 71.3% lower risk of death for dostarlimab than for paclitaxel plus doxorubicin and for pegylated liposomal doxorubicin, respectively; median survival was not reported. For PFS, the PHA was violated (P = 0.014). As such, the accelerated-failure-time model with Weibull distribution was fitted to the data. Despite the accelerated-failure-time model, no difference was noted between dostarlimab and carboplatin plus paclitaxel in PFS.19

Table 31. Summary of OS and PFS Before and After Matching (MAIC Report 3).

Table 31

Summary of OS and PFS Before and After Matching (MAIC Report 3).

The weighted ORR results for dostarlimab compared with paclitaxel or doxorubicin using the data from the McMeekin et al. (2015)56 study and carboplatin plus paclitaxel using data from the Mazgani et al. (2008)58 study are summarized in Table 32. For the comparison of dostarlimab with paclitaxel or doxorubicin in McMeekin et al. (2015),56 dostarlimab was favoured over paclitaxel and over doxorubicin using data from the McMeekin et al. (2015)56 study (OR = 0.202; 95% CI, 0.116 to 0.352). There was no difference between dostarlimab and paclitaxel plus carboplatin using data from the Mazgani et al. (2008)58 study.19

Table 32. Summary of ORR Before and After Matching (MAIC Report 3).

Table 32

Summary of ORR Before and After Matching (MAIC Report 3).

SAEs were only compared for the GARNET and McMeekin et al. (2015)56 studies and are summarized in Table 33. The results demonstrate no difference in SAEs between dostarlimab and paclitaxel or doxorubicin.

Table 33. Summary of SAEs Before and After Matching (MAIC Report 3).

Table 33

Summary of SAEs Before and After Matching (MAIC Report 3).

Inverse Probability Treatment Weighting

Description of IPTW Analyses

In addition to the 3 reports of MAIC analyses, the sponsor submitted 3 IPTW and propensity score matching (PSM) analyses to compare clinical outcomes in patients who received dostarlimab in GARNET with patients who received other treatments for advanced or recurrent EC, as follows19:

  • 1 analysis comparing IPD from GARNET with real-world patients receiving current treatment paradigms in a US real-world data cohort, using data from the Flatiron Health database (hereafter called IPTW report 1).
  • 1 analysis comparing IPD from GARNET with a GARNET-like RWE cohort in the UK from the NCRAS database (the base-case RWE cohort described in the MAIC section, hereafter called IPTW report 2).
  • 1 analysis comparing GARNET IPD with IPD from the doxorubicin arm of the ZoptEC trial, identified using the same SLR that was used to inform comparators for the previously described MAICs (hereafter called IPTW report 3).
Methods of ITPW Analyses
Objectives

The aim of each analysis was to compare clinical outcomes of survival (OS) in patients treated with dostarlimab from GARNET with OS in patients treated with current treatment paradigms for advanced or recurrent EC, based on external control cohorts from RWE databases and 1 clinical trial using IPTW methods. Additional objectives across the analyses included comparisons of outcomes of PFS, response (ORR, DOR), time on treatment (DoT, TTNT), HRQoL, and AEs, although these were not conducted for each comparison.19

Study Selection Methods

Study selection methods varied in the 3 sponsor-submitted IPTW reports. The index trial in all cases was based on IPD from IA-2 for cohort A1 of part 2B of the phase I, single-arm GARNET trial (dMMR or MSI-H EC cohort; n = 129). The selection characteristics used to develop the GARNET-like cohorts for the IPTW analyses for IPTW reports 1 and 3 are summarized in Table 34. The selection criteria for IPTW report 2 (using data from the NCRAS database) are summarized in Table 20.19

In IPTW report 1, no SLR was conducted to identify relevant comparator studies. Instead, an external real-world control cohort was created, leveraging data from the Flatiron Health database, a US nationwide, longitudinal, demographically and geographically diverse database derived from electronic health record data from more than 280 cancer clinics. Patients with recurrent or advanced EC who had progressed on or after no more than 2 prior lines of systemic chemotherapy for advanced or recurrent disease, with at least 1 being platinum-based therapy, were identified from the Flatiron Health Endometrial Cancer Analytic Cohort. The inclusion and exclusion criteria for the real-world retrospective cohort study were divided into 2 categories: criteria that were used to create the Flatiron Health Endometrial Cancer Analytic Cohort (n = 4,264); and additional criteria (adult age; no more than 2 prior lines of therapy, including at least 1 line of platinum-based chemotherapy [prior hormone therapy was allowed but did not count toward line of therapy]; additional line of therapy after platinum therapy for advanced or recurrent EC, and an ECOG Performance Status of 0 or 1 at index) (n = 185 after applying all criteria).19

In IPTW report 2, no SLR was conducted to identify relevant comparator studies. Instead, data from NCRAS were used, as previously described, for the MAIC analyses (refer to Table 20). The initial RWE cohort base case (N = 999) was used for the IPTW analysis, with patients with an ECOG Performance Status of 0 or 1 included as a scenario analysis (N = 501). There were a few minor differences between patient-level data for the RWE cohort and aggregated data for the same cohort used previously to conduct the MAICs, including differences in age and race/ethnicity, although these changes would not likely affect the results.19

In IPTW report 3, in consultation with clinicians and payer advisors, doxorubicin was identified as 1 of the most used chemotherapies in EC patients who progressed on platinum-based treatments. A clinical trial investigating doxorubicin (ZoptEC) was identified from the same SLR that was used to identify relevant comparator studies for the MAIC comparing data from GARNET with that from published literature. ZoptEC was a phase III, open-label, randomized, active-controlled, international study evaluating the efficacy and safety of AEZS-108 and doxorubicin in advanced or recurrent EC patients who received 1 platinum plus taxane combination regimen as a first-line treatment and progressed. The trial was discontinued in 2017 as it did not achieve its primary or secondary end points. The transfer of the IPD for the doxorubicin arm (N = 255) of this trial was negotiated by the sponsor. Key differences in clinical criteria between the GARNET and ZoptEC trials included prior anti-cancer regimens (up to 2 prior lines in GARNET versus 1 in ZoptEC), biomarker status (dMMR or MSI-H in GARNET versus not evaluated in ZoptEC), and ECOG Performance Status (≤ 1 in GARNET versus ≤ 2 in ZoptEC). Although dMMR or MSI-H status was not known in ZoptEC, as previously noted, its influence as a prognostic or treatment-effect modifier in advanced or recurrent EC is unknown. Thus, it was assumed that the GARNET and ZoptEC patients had comparable molecular profiles.19

Table 34. Selection Criteria for GARNET-Like Subgroups and for Relevant Comparator Studies for Sponsor-Submitted IPTW Analyses.

Table 34

Selection Criteria for GARNET-Like Subgroups and for Relevant Comparator Studies for Sponsor-Submitted IPTW Analyses.

The primary outcome of each analysis was OS, which was previously defined for the GARNET trial and the NCRAS RWE cohort (Table 21). In the Flatiron database cohort (IPTW report 1), OS was defined as the interval between the index date and the date of death, with the index date defined by the initiation date of therapy after post-platinum therapy (could consist of multiple lines of platinum therapy). OS was the only outcome for the IPTW analysis comparing GARNET with the NCRAS cohort (IPTW report 2). In the ZoptEC trial (IPTW report 3), OS was defined as time from date of the first dose of study treatment to the date of death by any cause.19

Other outcomes in the analyses using the comparison with the Flatiron database included TTD, previously defined for GARNET and icalculated as the time between the start date and end date of the index treatment, or censoring if the patient died, was lost to follow-up, or still on-therapy in the Flatiron database, as well as descriptive analyses of DoT and TTNT. In the comparison with the ZoptEC trial (IPTW report 3), additional outcomes included19:

  • PFS, calculated as time from the first dose to the previous date of assessment of progression or death by any cause in the absence of progression based on the time of first documentation of disease progression
  • ORR, defined as the proportion of patients that achieved BOR of CR or PR, per RECIST 1.1
  • DOR, defined as time from the first documentation of overall response leading to a confirmed CR or PR, when confirmation is required by RECIST 1.1, until time of the first documentation of overall response that included disease progression or death
  • TTD in HRQoL, defined as time from the first cycle to the earliest time with a decrease of at least 10 points (deterioration) from baseline in the global HRQoL; however, no rationale for this threshold was provided
  • AEs, including TEAEs, SAEs.
IPTW Analysis Methods

The method of each IPTW analysis varied. A summary of the analysis methods in each MAIC is provided below.

GARNET Versus Flatiron Cohort Methods (IPTW Report 1)

Five patients in GARNET who received anti-PD-L1/2 therapies after dostarlimab were excluded because of differences in availability across countries. As such, the analysis set from the GARNET trial was the safety cohort (n = 124), as was the efficacy analysis dataset (n = 103). After the application of inclusion and exclusion criteria to the 4,564 patients from the Flatiron Health Endometrial Cancer Analytic Cohort, the final study population of the real-world retrospective cohort study was 185 patients.19

A feasibility assessment was conducted to determine whether the retrospective real-world cohort was suitable for use as an external control arm to the GARNET dMMR or MSI-H EC cohort. Feasibility was based on assessments of data quality and information availability, consideration of sample size, availability of key prognostic factors and outcomes, and comparability with the GARNET trial. Overall, the feasibility assessment revealed that the definitions of all prognostic variables were comparable between cohorts; however, there were noted differences in reporting of ECOG Performance Status, BMI, number of prior anti-cancer regimens, prior surgery, and MMR or MSI-H status. Notably, MMR or MSI status was not known for most patients, and there were concerns about the under-reporting of prior surgeries in the Flatiron cohort. Based on the results of the feasibility assessment, a decision was made to proceed with the analysis.19

Prognostic variables in EC were identified and validated in accordance with the methods described previously for the initial RWE cohort MAIC. The prognostic factors used as covariates in the models included race, age, ECOG Performance Status, histology, FIGO stage, BMI, disease grade, number of prior platinum-based regimens, and prior surgery.19

The KM method was used to describe the distribution of OS and TTD by cohort. Survival curves were presented graphically. Median OS (95% CI) and estimated survival probabilities at specific time points were reported by cohort. The HR for OS and its 95% CI for the cohort variable (GARNET versus real-world cohort) were calculated from a Cox proportional hazard model in which cohort was included as a single covariate. The corresponding P value was also reported. The PHA was checked graphically by log-cumulative hazard plots for the covariate and by Schoenfeld residual plots. To assess whether the PHA was met, an interaction between time and the study variable was added to the Cox models as a time-dependent covariate.19

Covariate-adjusted comparisons of OS and TTD between cohorts were performed using regression modelling (only for OS), IPTW, and PSM. Propensity scores for each patient in the real-world control cohort were estimated as a patient’s predicted probability of being assigned to the GARNET cohort, which was estimated from a logistic regression model. The propensity score model included the key prognostic variables as covariates.19

Propensity scores for each patient were estimated from a logistic regression model as a patient’s predicted probability of being assigned to the GARNET cohort. Variable selection in the propensity score model considered clinical relevance (determined by key opinion leaders and medical experts), frequency of reporting in the literature, and strength of association with OS. Two propensity score models were built19:

  • A “lean” propensity score model that only included a small number of the most relevant of the variables; namely, histology, grade of disease at initial EC diagnosis, ECOG Performance Status, and number of prior platinum-based therapies in the advanced or recurrent setting.
  • A propensity score model that included all variables identified except prior surgery for EC.

The overlap of the distribution of the propensity scores across the GARNET cohort and real-world control cohort was assessed with a histogram of the estimated propensity scores in the 2 cohorts. PSM formed pairs of subjects from the GARNET and the real-world cohorts, such that matched subjects had similar propensity score values. To account for the imbalance in sample size between the 2 cohorts, a 1:2 matching ratio was used, where 2 patients from the real-world cohort were matched to each patient from the GARNET cohort. The matching was based on the greedy nearest neighbourhood matching without replacement, using a caliper width of 0.2. After matching, OS and TTD were compared between cohorts in the matched sample. This included the calculation of KM curves and estimation of median survival and corresponding 95% CIs.19

Adjusted HRs were obtained from multivariable Cox regression models, and included the prognostic variables as covariates in addition to the cohort variable. The same 2 sets of covariates as in the propensity score models were considered in the regression models.19

IPTW was performed using weights from estimated propensity scores. Weights were calculated from propensity scores so that resulting estimates referred to the average treatment effect (ATE). To assess balance in baseline characteristics between cohorts before and after weighting and matching, absolute standardized differences were calculated for each covariate, in which the standardized difference was defined as the difference in means or proportions divided by the pooled SD. Using the IPTW method, OS and TTD were compared between the cohorts in the weighted sample. This included the creation of weighted KM curves and the estimation of weighted median survival and corresponding CIs. A weighted Cox regression model for OS was also fitted to estimate adjusted HRs for the cohort variable.19

GARNET Versus NCRAS RWE Cohort Methods (IPTW Report 2)

In the analyses comparing GARNET with the RWE cohort from NCRAS conducted to assess balance in baseline characteristics between cohorts before weighting, absolute standardized differences were calculated for each covariate, in which the standardized difference is defined as the difference in means or proportions divided by the pooled SD. Statistical differences in baseline characteristics between the 2 treatment cohorts were evaluated using t-tests for continuous variables and chi-square tests for categorical outcomes.19

As above, the KM method was used to describe median (95% CI) OS and estimated survival probabilities by cohort. The HR for OS and its 95% CI for the cohort variable (GARNET versus real-world cohort) was calculated from a Cox proportional hazard model that included cohort as a single covariate. The PHA was checked graphically with log-cumulative hazard plots for the covariate and plots of Schoenfeld residuals for the treatment variable over time. In addition, Schoenfeld global tests that evaluated independence between Schoenfeld residuals and time were also performed.19

Covariate-adjusted comparisons of OS between cohorts were performed using IPTW based on the propensity score. Propensity scores for each patient were estimated as a patient’s predicted probability of being assigned to the GARNET cohort, which was estimated from a logistic regression model. The propensity score model included the key prognostic variables, identified following the method previously described for the RWE cohort MAIC, as covariates.19

Three different propensity score models were fitted. The sets of covariates used in the 3 propensity score models were identical to the sets of matching variables that defined the 3 MAIC scenarios described previously. The overlap of the distribution of propensity scores across the GARNET trial cohort and the RWE cohort was assessed with a histogram distribution graph of propensity scores across cohorts.19

Adjustment for potential imbalances in relevant prognostic variables between cohorts was done with the IPTW method. In the principal analysis, weights were calculated from propensity scores so that resulting estimates referred to the ATE. As a secondary analysis, weights were calculated from propensity scores so that resulting estimates referred to the ATE for the controls. The use of the ATE for controls perspective was motivated by the fact that this approach was similar to the weighting approach previously used in the MAIC, where each patient in the control cohort was assigned a fixed weight of 1.19

Using the IPTW method, OS was compared between the cohorts in the weighted sample. This included the creation of weighted KM curves and the estimation of weighted median survival and corresponding CIs. In the weighted KM curves, numbers at risk represent the sums of individual weights of patients at risk, rather than the number of patients at risk. A weighted Cox regression model was also fitted to estimate adjusted HRs (and 95% CIs, P values) for the cohort variable.19

GARNET Versus ZoptEC Methods (IPTW Report 3)

To facilitate a comparison between the single-arm GARNET trial and control patients using doxorubicin in the ZoptEC RCT, IPD from GARNET and ZoptEC were merged to create a comparator control arm.19

To make the baseline characteristics between the 2 trials comparable before statistical analysis, exclusion criteria were applied to each trial. Patients in the ZoptEC comparator trial who would have been ineligible in GARNET were removed before analysis if such criteria were easily determined from the dataset. Patients were excluded from the ZoptEC trial if they had a follow-up longer than 36 months, and patients were excluded if they did not have an ECOG Performance Status score of 0 or 1. Patients were excluded from the GARNET trial if they had previously received more than 1 prior platinum-based therapy. The main analysis dataset included 92 patients from GARNET and 233 patients from ZoptEC, and was used for OS, PFS, and ORR.19 A summary of datasets in the analysis is provided in Table 35.

A sensitivity analysis that included all patients from the GARNET and ZoptEC trials was conducted to ensure that the removal of patients did not violate the positivity or exchangeability assumption for the IPTW method. The positivity assumption can be empirically verified by showing there is a positive probability of each treatment for each set of covariates, whereas the exchangeability assumption assesses the potential impact of unmeasured confounding on the results using the methods of Mortimer et al. (2005)59 and Brumback et al. (2004),60 respectively. Additional sensitivity analyses included 1 that used the safety analysis dataset to assess whether this would affect the OS results; another of TTD in HRQoL was conducted using a 15-point decrease as the threshold.19

Table 35. Summary of Included Patients in Each Analysis Dataset.

Table 35

Summary of Included Patients in Each Analysis Dataset.

The main analysis dataset sample size — 233 for doxorubicin and 92 for dostarlimab — contained all available patients after the exclusion criteria were applied; thus, a sample size calculation was not applicable. However, a power analysis for OS was performed. If the HR for OS was greater than 0.65, the analysis would be underpowered; however, if the HR was less than or equal to 0.65, the analysis would be sufficiently powered. As with any statistical model using IPTW, power is decreased because IPTW inflates type I errors. Therefore, a stabilized IPTW was used, which is better suited in handling the inflation of type I errors and can also minimize any issues with statistical power.19

In the comparison between GARNET and the ZoptEC study, prognostic variables were identified and ranked with methods previously described for the RWE cohort published literature MAICs.19

A multivariable method with interaction terms for the assessment of effect modification was used for this analysis. For OS, Cox proportional hazards were used with covariates: treatment, candidate for effect modification (e.g., age or ECOG), and the interaction term (treatment × candidate of effect modification). If the P value for interaction was less than 0.05, the variable was considered an effect modifier. If effect modification was present, a subgroup analysis for each level of the effect modifier, in addition to the main analysis, was conducted.19

Because of the lack of randomization, there was a need to control for confounding bias at baseline, so an IPTW method was chosen. PSM was not chosen because there was a restricted number of patients in each arm coming from the clinical trial. A stabilized weight was estimated for each patient to reduce inflation of the type I error rate or to unintentionally increase statistical power. Once weighted, a Cox proportional hazards regression model with IPTW was used for the comparison of OS between dostarlimab and doxorubicin. KM analysis was used for PFS, DOR, and TTD in HRQoL. For binary outcomes such as an ORR, descriptive analyses were used. Specifically, the Clopper-Pearson method was used to calculate the 95% CI of the proportion. All assumptions, such as unmeasured confounding, positivity, and consistency, were checked in the validation of the IPTW method.19

Results of IPTW Analyses
Summary of Included Studies

Key study design characteristics and sources of heterogeneity of the studies included in the IPTW analyses are summarized in Table 36. There was considerable variation in the design of the 4 studies that were used in the IPTW analyses. The GARNET trial evaluated patients with dMMR or MSI-H EC, whereas the Flatiron, NCRAS, and ZoptEC studies included patients with advanced or recurrent EC, regardless of biomarker status. A total of 128 (69.2%) patients in the Flatiron database had unknown MMR or MSI-H status at index. There was also variation in the treatments included, with GARNET evaluating dostarlimab, the RWE cohorts evaluating the current treatment paradigms in advanced or recurrent EC, and the ZoptEC trial evaluating doxorubicin. Eligible patients typically had an ECOG Performance Status of 0 or 1; however, the ZoptEC trial enrolled patients with an ECOG Performance Status of 2, and ECOG status was unknown in most of the NCRAS RWE cohort. The line of therapy also varied across studies, with GARNET and the Flatiron RWE cohorts including patients with no more than 2 lines of therapy, 1 of which was a platinum doublet. The NCRAS real-world cohort used platinum doublet therapy as the key milestone regarding lines of therapy and did not analyze the lines of therapy before advanced or recurrent diagnosis. Moreover, the ZoptEC study only required 1 prior line of therapy.19 No consideration was noted regarding the differences in dosage, treatment or follow-up duration, or supportive care or cointerventions in the included studies.

Table 36. Key Study Characteristics in the GARNET and Comparator Studies.

Table 36

Key Study Characteristics in the GARNET and Comparator Studies.

GARNET Versus Flatiron Cohort (IPTW Report 1)

The baseline characteristics before weighting in the IPTW analysis of GARNET and the Flatiron RWE cohort are summarized in Table 37. In total, the GARNET safety population included 124 patients and the real-world cohort included 185 patients. There were 5 patients in GARNET (n = 129) not eligible for the comparison and excluded who received anti-PD-L1/2 after dostarlimab. In general, the populations of the GARNET trial and the Flatiron RWE cohort were relatively similar, except the GARNET trial had a higher proportion of White patients (75.0% versus 61.1%), endometrioid histology (66.1% versus 57.3%), FIGO stage I/II (43.5% versus 35.7%), and prior surgery (89.54% versus 56.2%). There was a substantial difference in disease grade between the GARNET and Flatiron cohorts because of the large amount of missing data for grade at diagnosis in the Flatiron cohort (38.4%). Comparability of the following variables between the 2 cohorts was limited because of discrepancies in variable definitions or data under-reporting of surgery and radiation in the Flatiron database: prior surgery for EC, history of radiation therapy before second-line therapy, and duration of the platinum- and progression-free interval at second line (because of the unavailability of response data in Flatiron, the platinum-free interval was measured differently in Flatiron and GARNET). The most frequent first-line regimen in the Flatiron cohort was carboplatin plus paclitaxel, received by the majority of patients (72.4%). During second-line treatment, the top 3 regimens were pegylated liposomal doxorubicin (17.8%), carboplatin plus paclitaxel (15.1%), and carboplatin plus docetaxel (9.7%). Only 105 patients received third-line therapy, with the most common being pegylated liposomal doxorubicin (15.2%), bevacizumab (9.5%), and carboplatin plus paclitaxel (7.6%).19

GARNET Versus NCRAS RWE Cohort (IPTW Report 2)

The baseline characteristics before weighting of the IPTW analysis of GARNET and the NCRAS RWE cohort are summarized in Table 37. In total, the GARNET cohort included 129 patients and the RWE base-case cohort included 999 patients. Patients in the GARNET trial were younger (51.2% versus 44.5% < 65 years). Overall, there were key differences in most characteristics of the patient populations; compared with the NCRAS cohort, GARNET had fewer White patients (76.0% versus 84.2%), and a greater proportion of patients with endometrioid histology (69.8% versus 42.4%), FIGO stage I and II disease (44.2% versus 22.1%), an ECOG Performance Status of 0 (42.6% versus 32.0%) or 1 (57.4% versus 18.1%), and a disease of grade 1 and 2 (67.4% versus 27.4%). The number of prior platinum-based therapies in the advanced or recurrent setting was 1 for all patients in the NCRAS RWE cohort, whereas it was 0 or 2 or more for 1.6% and 13.2% of patients in the GARNET cohort, respectively.19

GARNET Versus ZoptEC (IPTW Report 3)

The baseline characteristics before weighting of the IPTW analysis of GARNET and the ZoptEC trial are summarized in Table 37. The sponsor noted that the baseline characteristics were similar enough to warrant comparison. There were fewer White patients in GARNET than in the ZoptEC trial (76.0% versus 94.1%), and more patients with FIGO stage III and IV disease (85.3% versus 36.9%). The proportion of patients with endometrioid histology was similar. Comparability of some key variables, including BMI, disease grade, number of prior platinum therapies, and prior surgery, was limited because of a lack of reported data.19

Table 37. Baseline Characteristics of Patients by Cohort Before Weighting.

Table 37

Baseline Characteristics of Patients by Cohort Before Weighting.

Results

GARNET Versus Flatiron Cohort (IPTW Report 1)

Propensity scores for each patient were estimated from a logistic regression model as a patient’s predicted probability of being assigned to the GARNET trial. Results from the lean and full propensity score models fitted on the GARNET trial compared with the Flatiron cohort are summarized in Table 53. The overlap of the distribution of the propensity scores was assessed with a histogram distribution graph of propensity scores across cohorts (refer to Figure 12 and Figure 13 in Appendix 4). The lean model demonstrated greater overlap than the full model, so only the lean model was used.19

The distributions of baseline and prognostic factors considered for ITC analyses for GARNET and the Flatiron cohort after lean model IPTW and PSM are summarized in Table 54. Before and after matching, baseline and prognostic variables of race, FIGO, and BMI were different in GARNET and the Flatiron cohort. The matched GARNET cohort had a sample size of 93, and the matched real-world cohort had a sample size of 103.19

Results for OS and TTD before and after IPTW and PSM are summarized in Table 38. Median OS after IPTW was not estimable in GARNET (95% CI, 15.4 to not estimable) and was 13.1 (95% CI, 8.3 to 15.9) in the Flatiron cohort. Median OS after PSM was not estimable in GARNET (95% CI, 17.1 to not estimable) and was 15.9 (95% CI, 10.4 to 33.2) in the Flatiron cohort. Survival rates were higher for dostarlimab than for the Flatiron current treatment paradigm cohort; after IPTW, 24-month OS rates were 0.529 (95% CI, 0.367 to 0.667) in the GARNET cohort and 0.338 (95% CI, 0.262 to 0.415) in the real-world cohort. After PSM, 24-month OS rates were 0.536 (95% CI, 0.387 to 0.664) in the GARNET cohort and 0.410 (95% CI, 0.305 to 0.512) in the real-world cohort. Although the point estimates differ, 95% CIs overlapped, indicating no difference in survival rates.19

Cox proportional hazards regression models were fitted for OS to examine the association between cohort (GARNET versus Flatiron) and the risk of death. The PHA was checked graphically with a log-cumulative hazard plot and with a plot of Schoenfeld residuals for the treatment variable over time. In addition, an interaction between time and study was added to the Cox model as a time-dependent covariate. If the interaction term was not statistically significant, it was reasonable to assume that the PHA was valid.19

Unadjusted Cox proportional hazards regression models for both GARNET and the Flatiron cohort suggested that GARNET patients had a lower risk.(HR = 0.447; 95% CI, 0.305 to 0.653). When interaction for study and time were included, there was no difference in risk of death between studies (HR = 0.989; 95% CI, 0.927 to 1.055). For the model fitted after IPTW, the interaction term was significant when only the study variable was included (HR = 0.559; 95% CI, 0.385 to 0.812) but was not statistically significant when study and time variables were included. After PSM, the interaction terms were not statistically significant (HR for study = 0.653; 95% CI, 0.413 to 1.033; HR for study and time = 1.030; 95% CI, 0.955 to 1.111) (Table 55), so it was reasonable to assume that the PHA holds.19

Results from the multivariable Cox proportional hazards model for OS where 2 sets of covariates were adjusted are summarized in Table 56. The interaction term of study and time was not statistically significant in either adjustment scenario, which indicated that the PHA was valid for study. The results suggested that the study variable favoured the GARNET trial for risk of death after adjustment for the covariates in both adjustment scenarios. Table 57 shows the Cox proportional hazards model results for OS adjusted for covariates and propensity scores. For the study variable, the HRs adjusted for propensity score and for propensity score and covariates were consistent.19

For the exploratory outcome of TTD before and after IPTW and PSM, the results were congruent with the primary analysis of OS, with higher TTD rates at 24 months in the GARNET trial than in the Flatiron database (Table 38). The TTD rate after IPTW was 0.480 (95% CI, 0.319 to 0.625) in GARNET and 0.149 (95% CI, 0.081 to 0.237) in the real-world cohort. After PSM, the TTD rate at 24 months was 0.397 (95% CI, 0.272 to 0.520) in GARNET and 0.173 (95% CI, 0.084 to 0.287) in the real-world cohort; however, the overlap in the 95% CIs indicated that there was no difference.19

Table 38. OS Before and After IPTW and PSM — GARNET Versus Flatiron Real-World Cohort (IPTW Report 1).

Table 38

OS Before and After IPTW and PSM — GARNET Versus Flatiron Real-World Cohort (IPTW Report 1).

GARNET Versus NCRAS RWE Cohort (IPTW Report 2)

Baseline characteristics of the GARNET and NCRAS base-case cohorts after IPTW from 3 propensity score models are summarized in Table 58. Propensity score model 1 and 2 removed patients with unknown histology and patients with 0 or at least 2 prior platinum-based therapies from the GARNET cohort, which resulted in a sample size of 109 for the GARNET cohort; propensity score model 3 only removed patients with unknown histology from the GARNET cohort, which resulted in a sample size of 128 for the GARNET cohort. The standardized differences for variables used in the propensity score models were smaller after IPTW, compared with unweighted differences.19

Prior to IPTW, median OS could not be estimated for the GARNET cohort and was 10.3 months (95% CI, 9.2 to 11.1) for the RWE base-case cohort. The survival rates remained higher in the GARNET than in the RWE cohort during the study. At month 24, the survival rate was 0.578 (95% CI, 0.450 to 0.686) in the GARNET cohort and 0.215 (95% CI, 0.186 to 0.244) in the RWE base-case cohort. OS after IPTW that compared ATE weights for GARNET with the NCRAS RWE base-case cohort is summarized in Table 39. After IPTW, results for all propensity score models were consistent, with a greater median OS for dostarlimab than for the current treatment paradigm in the NCRAS RWE cohort (median OS was not evaluable at follow-up in GARNET and was 10.3 for all NCRAS models). Across all models after weighting, survival rates at 24 months ranged from 0.517 (95% CI, 0.326 to 0.678) to 0.606 (95% CI, 0.440 to 0.736) for GARNET and from 0.216 (95% CI, 0.187 to 0.246) to 0.218 (95% CI, 0.189 to 0.248) for the NCRAS RWE cohort. Using ATE for controls weights, results were consistent with ATE. Results of sensitivity analyses in patients with a ECOG Performance Status of 0 or 1 were consistent with the primary analysis.19

The HR for the cohort variable (GARNET versus real-world cohort) were calculated from a Cox proportional hazards model that included cohort as a single covariate. In all models, the HRs ranged from 0.310 (95% CI, 0.271 to 0.355) to 0.438 (95% CI, 0.386 to 0.498) (Table 39).19

The PHA was checked graphically with means of log-cumulative hazard plots for the covariate and plots of Schoenfeld residuals for the treatment variable over time. Both the graphs and the tests suggest that the PHA was valid for the unadjusted Cox models, as well as for the weighted Cox models based on weights calculated from propensity score model 3. The weighted Cox models based on weights calculated from propensity score models 1 and 2 do not seem to meet the PHA.19

Table 39. OS Before and After IPTW — GARNET Versus RWE Base-Case Cohort Propensity Score Models (ATE; IPTW Report 2).

Table 39

OS Before and After IPTW — GARNET Versus RWE Base-Case Cohort Propensity Score Models (ATE; IPTW Report 2).

GARNET Versus ZoptEC (IPTW Report 3)

The median OS of dostarlimab was not reached (95% CI, 17.150 to not reached), whereas the median OS for doxorubicin was 11.039 months (95% CI, 9.988 to 13.010). A naive HR was calculated using the lower-bound CI for dostarlimab, resulting in an unadjusted HR of 0.644. The main analysis was performed using a Cox proportional hazards model with stabilized IPTW to estimate the HR for OS (dostarlimab versus doxorubicin). A proportionality test was conducted to ensure that the PHA was not violated. As the P value was greater than 0.05, the PHA was supported, and the adjusted analysis used a Cox proportional hazards model with stabilized IPTW. KM curves for OS before and after adjustment are shown in Table 37. After adjusted stabilized IPTW, results of the Cox proportional hazards model suggested that dostarlimab was favoured over doxorubicin for OS (HR, 0.409 [95% CI, 0.277 to 0.605]). Survival time was significantly longer for dostarlimab, with median OS not reached (95% CI, 18.004 to not reached); for doxorubicin, median OS was 11.170 months (95% CI, 9.988 to 13.076).19

Kaplan-Meier estimates of OS in the stabilized IPTW population.

Figure 9

KM Curves for OS Without and With Adjusted Stabilized IPTW (IPTW Report 3).

A sensitivity analysis was conducted using the full safety analysis population from the GARNET (n = 129) and ZoptEC (n = 249) trials. Results were consistent with the main analysis, with dostarlimab favoured over doxorubicin for OS (HR, 0.403 [95% CI, 0.280 to 0.581]).19

Critical Appraisal of Indirect Evidence

Six ITC reports were submitted by the sponsor: 3 MAIC reports and 3 IPTW reports. The choice to conduct unanchored MAICs and IPTW analyses was justified by the lack of a common comparator. In all cases, the GARNET trial was used as the index trial. In 1 MAIC and 1 IPTW analysis, the comparator data were identified with a SLR, so there is a low risk of selection bias. For the remaining MAICs and IPTWs, no SLR was conducted to identify comparator studies. Instead, data from the UK NCRAS RWE database and the Flatiron Health database were used. As these were not selected using a systematic approach, there is a high risk of selection bias. It is not possible to know whether the results would have differed if data from a different database had been used.

There were important differences in the design of the comparator studies that limit the ability to draw strong conclusions about the efficacy of dostarlimab compared with other treatments. GARNET was a phase I, single-arm trial, whereas the comparators included real-world cohorts from the Flatiron electronic health records in the US and the NCRAS real-world database in the UK, published literature that consisted of retrospective and prospective studies, and a phase III RCT. Data analyzed retrospectively from databases and medical records are prone to unique biases (e.g., selection bias, confounding), compared with those collected from prospective, interventional studies (like RCTs and single-arm trials) that cannot be controlled for using MAICs or IPTWs.

The data-collection period and setting of the included studies varied, with enrolment for some beginning as far back as 2008 and for GARNET beginning in 2017. Some studies recruited internationally, whereas others recruited from single nations (Canada or US). There may be differences in clinical practice by region at various time points, although the direction of potential bias is unclear. Additionally, line of therapy definitions may not always be reflective of those used in clinical trials. GARNET included patients who had received no more than 2 lines of anti-cancer therapy for recurrent disease, whereas the NCRAS RWE database did not analyze lines of therapy before diagnosis of advanced or recurrent EC (all patients were receiving strictly second-line therapy). In the ZoptEC trial, patients only received 1 prior line of platinum doublet therapy. Follow-up duration also varied. Because the GARNET trial had an OS follow-up of less than 36 months in the published literature MAIC, the sponsor restricted the follow-up of each comparator study to no more than 36 months, and in the GARNET versus ZoptEC IPTW, patients with 36 months or more of follow-up were excluded. The comparative evidence is therefore limited to the longest follow-up for GARNET, for which survival data are still immature. Future comparisons after a longer follow-up may be more meaningful.

An important limitation of the MAICs and IPTWs is that in all comparator studies, MMR and MSI status was unknown for all or most patients. The indicated population for dostarlimab, according to the product monograph, is dMMR or MSI-H patients with advanced or recurrent EC. Further, the clinical experts consulted by CADTH for this review noted that MMR or MSI-H status has potential prognostic implications in these patients. It is therefore uncertain whether the comparator population in the MAIC and IPTW analyses would be eligible for treatment with dostarlimab, precluding strong conclusions about comparative efficacy. Furthermore, the comparators were based on those relevant to the advanced or recurrent EC population, rather than the dMMR or MSI-H subgroup; however, given the lack of advancement in funded treatments for advanced or recurrent EC overall, it may be reasonable that dMMR and MSI-H patients would receive similar options. The comparators included in the MAIC and IPTW were paclitaxel monotherapy, carboplatin plus paclitaxel, carboplatin plus liposomal doxorubicin, liposomal doxorubicin monotherapy, doxorubicin monotherapy, pegylated liposomal doxorubicin monotherapy, carboplatin monotherapy, and, because of limitations of the available data from RWE databases, the current treatment paradigm, which included a basket of possible treatments in advanced or recurrent EC. According to the clinical experts consulted by CADTH, these comparator treatments were relevant to Canadian clinical practice; however, 1 of the experts noted that MSI-H EC may be generally less responsive to platinum chemotherapy, although it is still used as first-line treatment in the absence of an alternative for dMMR or MSI-H patients. There were additional single-drug chemotherapies that could have been considered but were not included, such as gemcitabine and etoposide, because evidence for the efficacy of these treatments is lacking.

All comparisons investigated OS, an important outcome, according to the clinical experts consulted by CADTH for this review. Other outcomes were less frequently investigated, and those that are specifically important to patients (e.g., harms, HRQoL) were notably absent because comparative data were unavailable for those outcomes. There were some notable differences in outcome definitions, time of assessment, and follow-up duration across the comparators. Progression was not recorded in the NCRAS database, and TTNT and TTD were used as proxies, which may both over- and underestimate PFS. As such, strong conclusions cannot be made for this comparison outcome.

In the submitted MAIC analyses, an attempt to match patients based on inclusion and exclusion criteria was only conducted for the analysis versus published literature. In the MAIC of the NCRAS RWE cohort, patients met all of the applied inclusion criteria and none of the applied exclusion criteria for the GARNET trial; however, no information on this process was provided, and no matching of inclusion and exclusion criteria was done before weighting. An attempt to match inclusion and exclusion criteria was illustrated in the IPTW analysis with the Flatiron cohort, as well as in the IPTW and PSM comparisons with ZoptEC. Regardless of the scenarios or treatments, in the majority of cases, the ESS was greatly reduced, representing substantial losses to the precision of estimates. Thus, there was either considerable heterogeneity between GARNET and the RWE studies for the variables included in the weighting process, or the inclusion and exclusion criteria differed greatly in the studies, which is an important limitation of the relative treatment-effect estimates. In the absence of such evidence, the NICE Decision Support Unit considers the amount of bias in an unanchored MAIC likely to be substantial.61 For the primary outcome of OS, all results were associated with moderately to severely wide CIs, highlighting losses in precision and reducing the ability to draw strong conclusions.

In the IPTW analyses for the Flatiron and NCRAS RWE cohorts, propensity scores for each patient were estimated as a patient’s predicted probability of being assigned to the GARNET cohort, which was estimated from a logistic regression model. In the GARNET versus ZoptEC IPTW, exclusion criteria were applied to each trial in the analysis with the ZoptEC trial, followed by propensity scoring and weighting and a Cox proportional hazards model. One limitation of the IPTW technique is that it presumes all biases and confounding have been adjusted for in the model. To address this limitation, multiple PSMs were constructed in the analyses (particularly for Flatiron and NCRAS RWE cohorts) and the sets of covariates were defined based on expert opinion and results from the empirical assessment of effect modification and prognostic value of each covariate. However, it is still likely that there exist other confounders that were not observed and collected in the data. Moreover, in analyses of the Flatiron cohort and ZoptEC trial, the method of calculation was not reported, so the validity of the propensity scores could not be assessed. It was also unclear in all cases if any patients were excluded after propensity scoring.

The sponsors conducted a search of the literature and consulted with clinical experts to identify potential prognostic factors or treatment-effect modifiers in patients with advanced or recurrent EC. The key limitation of the MAICs is inherent in unanchored indirect comparisons, which assume that absolute outcomes can be predicted from the covariates (i.e., all effect modifiers and prognostic factors are accounted for in the model). This assumption is largely considered impossible to meet, according to the NICE Decision Support Unit technical guidance report on methods for population-adjusted indirect comparisons.61 The list of potential prognostic and treatment-effect modifiers was comprehensive and populated using appropriate methods; however, not all variables were available in each trial and, therefore, were not included in the models. Baseline characteristics were presented before and after weighting for all analyses. After the matching of baseline characteristics, the populations in all MAIC analyses were similar; however, in the IPTW analyses, some differences remained after IPTW and PSM, so the comparisons were not balanced for confounders and were not mutually randomizable populations. Aside from the known differences related to the availability of certain characteristics, such as ECOG Performance Status, prior lines of therapy, disease grade, and prior surgery, there were no additional concerns with the baseline characteristics after matching.

Overall, the phase I nature of the GARNET trial limits the ability to make definitive conclusions on comparative efficacy, given the short duration of follow-up resulting in immature data and the limited comparability with other studies because of differences in design. Moreover, because of the heterogeneity of the included populations, the comparisons conducted are not reflective of the Health Canada indication for dostarlimab; all comparator trials were not specific to dMMR or MSI-H patients as there was no comparative evidence identified for this population. The results of the MAICs and IPTWs were generally uncertain because of the clinical differences between populations, which resulted in reduced sample sizes after matching and the wider CIs. Moreover, it is highly unlikely that all prognostic factors and effect modifiers were accounted for, increasing the uncertainty in the results. As such, comparative efficacy in terms of improved survival must consider these limitations and interpretations must be made with caution, as results may not be generalizable to the indicated population. Last, outcomes important to patients, including improved HRQoL and reduced AEs, were not analyzed in the ITCs, so the comparative efficacy of dostarlimab for these outcomes remains uncertain.

Discussion

Summary of Available Evidence

The CADTH systematic review included 1 phase I trial that evaluated the efficacy and safety of dostarlimab in patients with dMMR or MSI-H recurrent or advanced EC that had progressed on or after prior treatment with a platinum-containing regimen. The GARNET trial (N = 129) is an ongoing, multi-centre, open-label, single-arm, phase I dose-escalation and cohort-expansion study of patients with recurrent or advanced solid tumours. This CADTH review focused on cohort A1, which aligned with the proposed Health Canada indication and the reimbursement request. All patients received dostarlimab through IV injection (500 mg every 3 weeks for cycles 1 to 4, and 1,000 mg every 6 weeks from cycle 5 onward) for up to 2 years or until disease progression or unacceptable toxicity, whichever occurred first. The co-primary outcomes were ORR and DOR, and secondary outcomes were OS, PFS, DCR, irPFS, irDCR, irORR, and irDOR. HRQoL was an exploratory outcome. AEs and irAEs were also monitored and reported. The statistical analysis plan of the GARNET trial specified 3 interim analyses. The focus of this CADTH review was IA-2, which was the most recent interim analysis available (median follow-up time = 16.3 months). IA-3 was planned for November 1, 2021, but data from this analysis were not submitted to CADTH until later in the review process so were not included as part of the main analysis. The IA-3 results were, overall, consistent with those reported in IA-2. For further details on IA-3, refer to Appendix 5.

In cohort A1 of the GARNET trial, the majority of enrolled patients with dMMR or MSI-H EC were White, had type II and grade 3 endometrioid tumours, had FIGO stage IV disease, had an ECOG Performance Status of 1, had received 1 or 2 previous lines of systemic therapy for recurrent or advanced EC, and had a median age of 64 (range = 39 to 84) years. Limitations of the GARNET trial included potential biases inherent to its single-arm, noncomparative trial design, which prohibits the ability to draw causal conclusions about the intervention and outcomes.

In the absence of comparative evidence, the sponsor submitted 6 reports of ITCs — 3 MAIC reports and 3 IPTW and PSM reports — that compared dostarlimab with currently available treatment regimens using RWE and data from the published literature. The primary end point for all comparisons was OS. Other outcomes included PFS, ORR, DOR, TTD, DoT, TTNT, TTD in HRQoL, and AEs; however, these were less frequently investigated, and outcomes specifically important to patients, such as HRQoL, were not assessed. There were important differences in the design of the comparator studies that limit the ability to draw strong conclusions about the efficacy of dostarlimab compared with other treatments. An important limitation of all analyses was that MMR and MSI-H status was unknown for all or most patients in the comparator trials, so it is uncertain whether the comparator populations in the ITC analyses would be eligible for treatment with dostarlimab; this creates further uncertainty about comparative efficacy.

Interpretation of Results

Outcomes

The achieved ORR and DOR (co-primary end points in cohort A1) were considered clinically meaningful by the clinical experts consulted by CADTH. The clinical experts felt that the ORR of 44.8% (95% CI, 35.0% to 54.8%) was both clinically meaningful and durable for this patient population. Although the median DOR was not reached, approximately 79% of dostarlimab responders had a DOR of at least 6 months, which the clinical experts noted was impressive compared with currently available treatments. The GARNET trial included no formal hypothesis or statistical significance testing; as such, point estimates with 95% CIs were reported to estimate the magnitude of treatment effect associated with dostarlimab. Given that the trial was not designed to detect differences in treatment effects across subgroups, no conclusions can be drawn from pre-specified subgroup results. OS and PFS were assessed as secondary outcomes in the GARNET trial; however, interpretation of time-to-event end points is limited in a single-arm trial. Because all patients in cohort A1 of the GARNET trial received the same treatment, it remains unclear if the observed survival rates were due to the natural history of the disease or treatment with dostarlimab. Another limitation was that the median OS was not reached at the time of IA-2. Although KM estimates were provided at different time points, these data may overestimate the efficacy of the treatment given the immaturity of the data.

According to the clinical experts consulted by CADTH and the registered-clinician groups that provided input for this submission, the tumour response outcomes, including the CR rate and DOR, achieved with dostarlimab in the GARNET trial were clinically meaningful and much higher than those observed with therapies currently used in this setting. According to the clinical experts, the CR and response rates achieved in the study population were excellent compared with the CR rates they see in clinical practice and with other historical immunotherapies. Although the results of the MAIC and IPTW analyses generally suggest that dostarlimab is favoured for OS over all the included comparators, there was significant uncertainty in the results based on the clinical heterogeneity of the included study populations, resulting in reduced sample sizes and treatment-effect estimates that had wide CIs.

The clinical experts emphasized the clinical relevance and importance of maintaining stable disease to prevent an otherwise fast decline in patients; for many, second-line therapy may be their last line of treatment. This view was echoed in the input provided by the patient-advocacy group, which highlighted tumour response, maintenance of response, delay in disease progression, and HRQoL as important treatment goals for patients. Although the clinical experts agreed that, based on the available evidence, it was not possible to conclude whether the antitumour activity expressed as responses would translate into PFS and OS benefit, they felt that the preliminary survival results from the trial (median PFS was 5.5 months [95% CI, 3.2 to NR] and median OS was not reached) were encouraging, and that the durable responses observed could potentially delay tumour progression and result in prolonged survival in this patient population.

Input received from the patient-advocacy and clinician groups, as well as the clinical experts consulted by CADTH, highlighted HRQoL as an important outcome and treatment goal for patients. The clinical experts consulted by CADTH noted that HRQoL in patients with advanced or recurrent EC is low and unstable, and they did not foresee worsening of HRQoL with dostarlimab, given its acceptable toxicity profile. However, conclusions could not be drawn from the HRQoL evidence from the GARNET trial, given its noncomparative, open-label trial design, the substantial decline over time in patients who completed questionnaires, the lack of statistical analyses, and the lack of a MID or definition of what constituted a clinically meaningful change from baseline in the trial population. The clinical experts and clinician groups anticipated that given the responses and manageable toxicities observed in the GARNET trial, dostarlimab would likely improve or at least maintain patients’ HRQoL; however, these expectations need to be confirmed in a randomized clinical trial.

Although patients in the GARNET trial were considered to be representative of patients in Canadian clinical practice, the clinical experts noted that study patients were slightly younger than the patients typically seen in clinical practice; still, they noted, results would be generalizable. The clinical experts anticipated that because of its acceptable safety profile, they would expect to see a benefit of treatment with dostarlimab, regardless of the number of previous lines of systemic therapy, in patients with dMMR or MSI-H EC. However, patients should not have previously been treated with other anti-PD-1 or anti-PD-L1 immunotherapies.

Harms

The single-arm, nonrandomized design of the GARNET trial made it a challenge to interpret the safety events attributable to dostarlimab, because all patients in cohort A1 received the same treatment. Almost all patients in cohort A1 experienced at least 1 TEAE (n = 123; 95.3%) and almost 50% of TEAEs were grade 3 or higher. The clinical experts consulted by CADTH noted that most TEAEs associated with dostarlimab are manageable with supportive care and are not life-threatening. Further, they noted that treatment discontinuation due to TEAEs was relatively rare. The prevalence of treatment toxicities was in line with what is expected with other immunotherapies. From the review of notable harms, the clinical experts noted that the immune-related toxicities seemed to be associated with dostarlimab treatment. Overall, death from AEs was minimal, and no TEAEs that led to death were considered to be treatment-related. The clinical experts agreed with clinician group input that the TEAEs observed with dostarlimab were, overall, acceptable and could be adequately managed in clinical practice. This was reflective of patients’ experience with dostarlimab reported in the patient input, which stated that, overall, they had few challenges dealing with side effects of dostarlimab. Furthermore, it was emphasized by the clinical experts consulted by CADTH that the toxicity associated with dostarlimab appeared favourable, compared with currently available chemotherapy options. Examples of side effects from chemotherapy that may be weaker with dostarlimab include alopecia, fatigue, peripheral neuropathy, neutropenia, and febrile neutropenia, according to the clinical experts. The sponsor-submitted ITCs reports did not assess safety outcomes.

Conclusions

One phase I, singe-arm, open-label trial (GARNET)18 provided evidence on the efficacy and safety of dostarlimab in adults with dMMR or MSI-H recurrent or advanced EC (cohort A1) that had progressed on or after prior treatment with a platinum-containing regimen. The clinical experts consulted by CADTH felt that the co-primary response outcomes of ORR and DOR observed in the trial were clinically meaningful and durable for this patient population and, in their opinion, were higher than what is observed with currently used second-line therapies in this setting. The trial results were based on an interim analysis, so it is possible that clinical benefit was overestimated and harms were underestimated. There was uncertainty around the magnitude of the clinical benefit, given the limitations inherent to the single-arm trial design. The trial data on important long-term outcomes were immature, and interpretation of OS will be confounded by the use of subsequent anti-cancer therapies. The clinical experts noted that a RCT would be needed to directly compare dostarlimab with currently available therapies in the second-line setting to accurately evaluate its efficacy in this patient population. In the absence of a direct comparison of dostarlimab with relevant treatment options, the sponsor submitted multiple ITCs. However, the CADTH critical appraisal of these analyses identified significant limitations with the submitted MAICs and IPTWs that restricted the ability to interpret the relative treatment-effect estimates obtained. Limitations of the ITCs included heterogeneity across study designs, high risk of confounding and effect modifiers, and uncertainty regarding the inclusion of dMMR or MSI-H in the comparator groups. The results for HRQoL, an outcome important to patients and clinicians, remained inconclusive because of the lack of statistical analysis, substantial decline in patients completing questionnaires over time, and the lack of a definition of what constituted a clinical meaningful change from baseline. The notable harms observed with dostarlimab, such as diarrhea and peripheral nephropathy, were considered manageable and consistent with other immunotherapies by the clinical experts and, in their opinion, appeared favourable when naively compared with currently available chemotherapy options. However, interpretation of the safety events attributable to dostarlimab was a challenge because all patients in cohort A1 received the same treatment. Overall, limitations of the single-arm design of the GARNET trial prohibited the ability to draw causal conclusions between the intervention and outcomes.

Abbreviations

AE

adverse event

ATE

average treatment effect

BICR

blind independent clinical review

BMI

body mass index

BOR

best overall response

CCO

Cancer Care Ontario

CI

confidence interval

CR

complete response

DCR

disease control rate

dMMR

deficient mismatch repair

DOR

duration of response

DoT

duration of treatment

EC

endometrial cancer

ECOG

Eastern Cooperative Oncology Group

EORTC QLQ-C30

European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30

EOT

end of treatment

EQ-5D-5L

EQ-5D 5-Levels

EQ-VAS

EQ-5D-5L visual analogue scale

ESS

effective sample size

FIGO

International Federation of Gynecology and Obstetrics

GOC

Society of Gynecologic Oncology of Canada

HR

hazard ratio

HRQoL

health-related quality of life

IA-1

first interim analysis

IA-2

second interim analysis

IA-3

third interim analysis

IHC

immunohistochemistry

IPD

individual patient data

IPTW

inverse probability treatment weighting

irAE

immune-related adverse event

irDCR

immune-related disease control rate

irDOR

immune-related duration of response

irORR

immune-related objective response rate

irPFS

immune-related progression-free survival

irRECIST

immune-related Response Evaluation Criteria in Solid Tumours

ITC

indirect treatment comparison

KM

Kaplan-Meier

MAIC

matching-adjusted indirect comparisons

MID

minimally important difference

MMR

mismatch repair

MMR-unk

unknown mismatch repair tumour status

MSI

microsatellite instability

MSI-H

microsatellite instability-high

MSS

microsatellite stable

MUHC

McGill University Health Centre

NCRAS

National Cancer Registration and Analysis Service

NGS

next-generation sequencing

NICE

National Institute of Health Care Excellence

NOC/c

Notice of Compliance with conditions

OH

Ontario Health

ORR

objective response rate

OS

overall survival

PD-1

programmed cell death protein-1

PD-L1

programmed death ligand-1

PD-L2

programmed death ligand-2

PFS

progression-free survival

PHA

proportional hazards assumption

PMCC

Princess Margaret Cancer Centre

PR

partial response

PSM

propensity score matching

RCT

randomized controlled trial

RECIST

Response Evaluation Criteria in Solid Tumours

RWE

real-world evidence

SAE

serious adverse event

SBHSC

Sunnybrook Health Sciences Centre

SCA

Saskatchewan Cancer Agency

SD

standard deviation

TEAE

treatment emergency adverse effect

TTD

time to treatment discontinuation

TTNT

time to next treatment

Appendix 1. Literature Search Strategy

Note that this appendix has not been copy-edited.

Clinical Literature Search

Overview

Interface: Ovid

Databases:

  • MEDLINE All (1946-present)
  • Embase (1974-present)

Note: Subject headings and search fields have been customized for each database. Duplicates between databases were removed in Ovid.

Date of search: November 3, 2021

Alerts: Bi-weekly search updates until project completion

Search filters applied: No filters were applied to limit the retrieval by study type.

Limits:

  • Conference abstracts: excluded
Table 40. Syntax Guide.

Table 40

Syntax Guide.

Multi-Database Strategy

  1. (dostarlimab* or jemperli* or P0GVQ9A4S5 or P-0GVQ9A4S5 or UNIIP0GVQ9A4S5 or UNIIP-0GVQ9A4S5 or TSR042 or TSR-042 or WBP285 or WBP-285 or GSK4057190 or GSK-4057190 or ANB011 or ANB-011).ti,ab,kf,ot,hw,nm,rn.
  2. 1 use medall
  3. *dostarlimab/
  4. (dostarlimab* or jemperli* or TSR042 or TSR-042 or WBP285 or WBP-285 or GSK4057190 or GSK-4057190 or ANB011 or ANB-011).ti,ab,kf,dq.
  5. 3 or 4
  6. 5 use oemezd
  7. (conference review or conference abstract).pt.
  8. 6 not 7
  9. 2 or 8
  10. remove duplicates from 9

Clinical Trials Registries

ClinicalTrials.gov

Produced by the US National Library of Medicine. Targeted search used to capture registered clinical trials.

[Search terms – dostarlimab or Jemperli]

WHO ICTRP

International Clinical Trials Registry Platform, produced by WHO. Targeted search used to capture registered clinical trials.

[Search terms – dostarlimab or Jemperli]

Health Canada’s Clinical Trials Database

Produced by Health Canada. Targeted search used to capture registered clinical trials.

[Search terms – dostarlimab or Jemperli]

EU Clinical Trials Register

European Union Clinical Trials Register, produced by the European Union. Targeted search used to capture registered clinical trials.

[Search terms – dostarlimab or Jemperli]

Grey Literature

Search dates: October 30 – November 3, 2021

Keywords: dostarlimab, Jemperli, endometrial cancer

Limits: None

Updated: Search updated before the meeting of CADTH pan-Canadian Oncology Drug Review Expert Committee (pERC).

Relevant websites from the following sections of the CADTH grey literature checklist Grey Matters: A Practical Tool for Searching Health-Related Grey Literature were searched:

  • Health Technology Assessment Agencies
  • Health Economics
  • Clinical Practice Guidelines
  • Drug and Device Regulatory Approvals
  • Advisories and Warnings
  • Drug Class Reviews
  • Clinical Trials Registries
  • Databases (free)
  • Health Statistics
  • Internet Search
  • Open Access Journals

Appendix 2. Excluded Studies

Note that this appendix has not been copy-edited.

Table 41. Excluded Studies.

Table 41

Excluded Studies.

Appendix 3. Description and Appraisal of Outcome Measures

Note this appendix has not been copy-edited.

Aim

To describe the following outcome measures and review their measurement properties (validity, reliability, responsiveness to change, and MID):

  • European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30) version 3.0
  • EQ-5D-5L version 2.0

Findings

Table 42. Summary of Relevant Secondary Outcomes and Their Measurement Properties.

Table 42

Summary of Relevant Secondary Outcomes and Their Measurement Properties.

European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30)

Description

The EORTC QLQ-C30 is a self-reported instrument designed to measure the physical, psychological, and social functions of patients with cancers.48 The EORTC QLQ-C30 consists of 30 items that are scored to create 5 multi-item functional scales, 3 multi-item symptom scales, 6 single-item symptom scales, and 2 global quality of life (QoL) scales (Table 43) Version 3.0 of the questionnaire is the most current version and has been in use since December of 1997.75,76 It is intended for use in the adult population only.48

Table 43. EORTC QLQ-C30 Scales.

Table 43

EORTC QLQ-C30 Scales.

Scoring

The EORTC QLQ-C30 uses a 1-week recall period to assess function and symptoms.76 Twenty-eight questions are scored on a 4-point Likert scale (1: not at all; 2: a little; 3: quite a bit; 4: very much). The 2 questions that make up the global HRQoL scale are scored on a 7-point Likert scale with anchors at 1 (“very poor”) and 7 (“excellent”).

Raw scores for each scale are computed as the average of the items that contribute to a particular scale.76 This scaling approach is based on the assumption that it is appropriate to provide equal weighting to each item that comprises a scale. There is also an assumption that, for each item, the interval between response options is equal (for example, the difference in score between “not at all” and “a little” is the same as “a little” and “quite a bit,” at a value of 1 unit). Each raw scale score is converted to a standardized score that ranges from 0 to 100 using a linear transformation, with a higher score reflecting better function on the function scales, higher symptoms on the symptom scales, and better HRQoL (i.e., higher scores simply reflect higher levels of response on that scale). Thus, a decline in score on the symptom scale would reflect an improvement, whereas an increase in score on the function and HRQoL scales would reflect an improvement. According to the EORTC QLQ-C30 scoring algorithm, if there are missing items for a scale (i.e., the patient did not provide a response), the score for the scale can still be computed if there are responses for at least one-half of the items. In calculating the scale score, the missing items are simply ignored — an approach that assumes that the missing items have values equal to the average of those items for what the respondent completed.

Psychometric Properties

The EORTC QLQ-C30 was originally validated in patients with lung cancer and head and neck cancer from various European and North American countries, as well as from Australia.62,77,78 The scales of the EORTC QLQ-C30 have been found to assess distinct components of HRQoL; distinguishing between patients with different performance status and degrees of weight loss, and responsive to change over time.62,77,78

A literature search was conducted to identify validation information of the EORTC QLQ-C30 in patients with EC and none were identified. Of note, there is a validated version of the EORTC QLQ that was developed specific for endometrial cancer: EORTC QLQ-EN24.79 The EORTC QLQ-EN24 was designed to assess disease and treatment-specific aspects of the HRQoL of patients with EC. A validation study of the Mexican-Spanish version of the EORTC QLQ-EN24 in 189 patients with EC included a brief validation of the QLQ-C30.80 The study confirmed the internal consistency and reliability of the QLQ-C30 and found that its convergent and discriminant validity (Cronbach alpha range = 0.77 to 0.89) was consistent with its original report.80

In a further validation study of the EORTC QLQ-C30, patients with breast cancer (n = 121), ovarian cancer (n = 111) lung cancer (n = 160) and a heterogenous group of other cancers (n = 121) completed the questionnaire before and on day 8 of chemotherapy.64 The item-domain correlations of the EORTC QLQ-C30 were not different across the primary tumour sites (i.e., breast, ovary or lung). The correlations for all items, except for item 5 (whether the responders needed help with eating, dressing, washing, or using the toilet) and the physical function domain (r = −0.3), were highly correlated within their own domain than with any other domains (r = −0.65 to 0.95). At day 8 of chemotherapy, the item-domain for item 5 and the physical function domain was 0.49 for the entire group and ranged from −0.38 for ovarian cancer to −0.55 for breast cancer. These higher values at day 8 suggest that item 5 was more relevant in the week after chemotherapy than before chemotherapy.64 Similarly, items asking about vomiting showed a higher correlation with domains for nausea/vomiting on day 8 after chemotherapy (r = 0.89) than before chemotherapy (r = 0.74). The questionnaire also demonstrated good internal consistency for most domains at baseline and at day 8 (Cronbach alpha > 0.70). However, values were < 0.70 at baseline and day 8 for role function (0.66 and 0.53, respectively) and cognitive function (0.63 and 0.58, respectively). An examination of the discrimination of the domain scores according to primary cancer site found that the mean scores at baseline were not significantly different from each other in all groups for emotional function, cognitive function, or nausea/vomiting. However, the mean scores for each of the other domains differed between the group, with patients with breast cancer tending to have better physical function, role function, social function, less fatigue, and pain and better global HRQoL. Patients with lung and ovarian cancer reported lower scores for all these domains, with patients with ovarian cancer reporting the lowest scores for all domains. After chemotherapy, many of the differences seen at baseline between the groups were no longer evident. At day 8, patients with ovarian cancer has the smallest magnitude of change, with no change in role function, social function and global HRQoL, while being the only cancer group reporting a significant improvement in pain.64

An analysis of data from a Canadian RCT of paclitaxel and cisplatin versus cyclophosphamide and cisplatin in the treatment of 153 patients with epithelial ovarian cancer found the EORTC QLQ-C30 adequately assessed the effect of expected toxicities on patients HRQoL during and after treatment.65 At baseline, before the initiation of treatment, there was close agreement in the “mild or none” category between the symptoms recorded on case report forms (CRFs) and paired EORTC QLQ-C30 questions. The greatest degree of agreement ranged between 0.80 (95% CI, 0.75 to 0.86) to 0.98 (95% CI, 0.92 to 0.99). The pairing of lethargy with Question 18, and mood with Question 22 were slightly weaker in agreement compared to the other pairs at 0.72 and 0.73, respectively. The weakest pairs were constipation with Question 16, and lethargy with Question 18 at 0.44 and 0.44, respectively. During treatment and at the end of cycles 3 and 6, all but 1 symptom and HRQoL pairs demonstrated marked agreement ranging from 0.71 to 0.93. The 1 exception was the pair assessing symptom hair loss and Question 42 with a degree of agreement of 0.50 and 0.37 at cycles 3 and 6, respectively. A regression model predicting global HRQoL scores based on baseline grades of the most frequently observed toxicities and scores corresponding to HRQoL question found that the questions related to motor weakness (question 12), anorexia (question 13), mood (question 24), gastrointestinal pain (question 40) and vomiting (question 15) explained 60% of the variance in baseline global HRQoL on the EROTC QLQ-C30. When patients were off chemotherapy, 78% of symptoms and HRQoL pairs had high level of agreement (> 0.80).

Minimal Important Difference

A literature search was conducted to identify the MID of the EORTC QLQ-C30 in patients with EC and none were identified. Below is a summary of the MID of the EORTC QLQ-C30 in patients with cancer in general.

Change in the EORTC QLQ-C30 may be interpreted in terms of small, moderate or large changes in HRQoL.67 A study of patients with breast cancer and small cell lung cancer estimated that a clinically relevant change in score on any of the EORTC QLQ-C30 scales to be 10 points.67 Using an anchor-based approach to estimate the MID in which patients who reported “a little” change (for better or worse) on the subjective significance questionnaire (SSQ) had corresponding changes on a function or symptom scale of the EORTC QLQ-C30 of approximately 5 to 10 points. Patients who reported a “moderate” change had corresponding changes in the EORTC QLQ-C30 of about 10 to 20 points, and those who reported being “very much” changed had corresponding changes of more than 20 points.

A Canadian study estimated the MID for the EORTC QLQ-C30 among 369 patients with advanced cancer, the most common cancer being breast cancer, followed by lung, prostate, gastrointestinal, renal cell, and other cancers.68 Patients completed the questionnaire at baseline and 1-month post-radiation. Using both an anchor- and distribution-based methods for improvement and deterioration, 2 anchors of overall health and overall HRQoL were used, both taken directly from the EORTC QLQ-C30 (questions 29 and 30) where patients rated their overall health and HRQoL themselves. Improvement and deterioration were categorized as an increase or decrease by 2 units to account for the natural fluctuation of patient scoring. With these 2 anchors, the estimated MIDs across all EORTC QLQ-C30 scales ranged from 9.1 units to 23.5 units for improvement, and from 7.2 units to 13.5 units for deterioration. Distribution-based estimates were closest to 0.5 SD.

EQ-5D-5L

Description

The EQ-5D-5L is a generic self-reported HRQoL outcome measure that may be applied to a variety of health conditions and treatments.49,69 The first 2 components of the EQ-5D-5L assesses 5 domains: mobility, self-care, usual activities, pain/discomfort and anxiety/depression. Each domain has 5 levels: no problem; slight problems; moderate problems; severe problems; and extreme problems. A descriptive system that classifies respondents (aged ≥ 12 years) based on the following 5 dimensions: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. The EQ-5D-5L has 5 possible levels for each domain and respondents are asked to choose the level that reflects their health state for each of the 5 domains resulting in 3,125 possible health states.70 The second component of the EQ-5D-5L part is a 20 cm visual analogue scale (EQ-VAS) that has end points labelled 0 and 100, with respective anchors of “worst imaginable health state” and “best imaginable health state.” Respondents are asked to rate their health by drawing a line from an anchor box to the point on the EQ-VAS which best represents their health on that day. Thus, the EQ-5D-5L produces 3 types of data for each respondent:

  1. A profile indicating the extent of problems on each of the 5 dimensions represented by a 5-digit descriptor, e.g., 15121, 33211;
  2. A population preference-weighted health index score based on the descriptive system; and
  3. A self-reported assessment of health status based on the EQ-VAS.
Scoring

The EQ-5D-5L index score is generated by applying a multi-attribute utility function to the descriptive system.71 Different utility functions are available that reflect the preferences of specific populations (e.g., US or UK). Scores less than 0 represent health states that are valued by society as being worse than dead, while scores of 0 and 1.00 are assigned to the health states ‘dead’ and ‘perfect health,’ respectively.

Psychometric Properties

A literature search was conducted to identify validation information of the EQ-5D-5L in patients with EC and none were identified. Below is a summary of the psychometric property of the EQ-5D-5L in patients with gynecological cancers.

Among 530 patients with cervical cancer in Taiwan, the EQ-5D-5L was found to be both a reliable and valid assessment of HRQoL.72 The interclass correlation of the EQ-5D-5L was 0.8, with Cohen kappa values for the different dimension ranging from 0.54 to 0.73. Its convergent and discriminant validities were examined using the EORTC QLQ-30 and the clinical indicators of the functional performance assessment using the Karnofsky Performance Scale (KPF) and disease status. The EQ-5D was strongly correlated with all EORTC QLQ-C30 functioning scales, and its index and VAS scores were higher for patients with higher KPS score and disease-free status.72

Among a group of 300 patients (mean age: 51.5 ± 11.5 years) with HPV-related cancer (i.e., head and neck cancer - 70%; cervical cancer - 13.4%; and nasopharyngeal cancer - 10%) in Indonesia, the EQ-5D-5L demonstrated good to excellent test-retest reliability across domains (ICCs: Mobility = 0.97; Self-care = 0.95; Usual activities = 0.79; Pain/discomfort = 0.84; Anxiety/depression = 0.82; EQ-5D visual analogue scale = 0.73) and good internal consistency (Cronbach alpha = 0.84).73 Construct and convergent validity of the EQ-5D-5L in this population was assessed by mapping its subscales to those of the EORTC QLQ-C30. Significant correlation between almost all the dimension of the Indonesian version of the EQ-5D-5L with mapped subscale of the EORTC QLQ-C30, including physical function, role function, fatigue and pain was observed. Only the mobility dimension of the EQ-5D-5L was not correlated with the social function subscale of the EORTC QLQ-C30.73

The responsiveness of the EQ-5D-5L was evaluated in a longitudinal study of 50 patients with cervical intraepithelial neoplasia in China. The EQ-5D-5L demonstrated only small to moderate responsiveness post-surgery. While scores for self-care and usual activities did not change, scores of mobility, pain/discomfort and anxiety/depression decreased by 0.003, 0.004 and 0.016, indicating improvement of these dimensions at follow-up.

Minimal Important Difference

A literature search was conducted to identify MID of the EQ-5D-5L in patients with EC and none were identified. Below is a summary of MID of the EQ-5D-5L in patients within the general population and in patients with other gynecological cancers.

A simulation-based approach using an instrumental-defined single-level transitions was used to estimate the MID of the EQ-5D-5L in the general population for each country-specific scoring algorithm. An estimated MID between 0.037 and 0.069 was determined for 6 countries (Canada, China, Spain, Japan, England, and Uruguay).81 The country-specific scoring algorithm were as follows Canada, 0.056 ± 0.011; China, 0.069 ± 0.007; Spain, 0.061 ± 0.008; Japan, 0.048 ± 0.004; England, 0.063 ± 0.013; and Uruguay, 0.063 ± 0.019. Differences in MID estimates reflect differences in population preferences, in valuation techniques used, as well as in modelling strategies. After excluding the maximum-valued scoring parameters, the MID estimates (mean ± SD) were as follows: Canada, 0.037 ± 0.001; China, 0.058 ± 0.005; Spain, 0.045 ± 0.009; Japan, 0.044 ± 0.004; England, 0.037 ± 0.008; and Uruguay, 0.040 ± 0.010.81

The MID of the of the EQ-5D-5L was determined in a longitudinal study of 50 patients with cervical intraepithelial neoplasia (CIN) in China using 3 methods: distribution-based, anchor-based, and instrument-defined methods.74 The MIDs, by anchor-based, instrument-defined, and anchor-based methods were 0.041, 0064 and 3.12, respectively. The MIDs estimated in this study; however, only represents truly meaningful change of HRQoL scores at the group level, not the individual level.74

Appendix 4. Detailed Outcome Data

Note that this appendix has not been copy-edited.

The results of OS from the subset of patients from IA-1 (N = 72) included in IA-2 was 26 deaths (36.1%) and 46 (63.9%) patients censored.

Table 44. KM Analysis of OS in Patients With dMMR or MSI-H EC (IA-1 Subset of Patients in IA-2 — Primary Efficacy Analysis).

Table 44

KM Analysis of OS in Patients With dMMR or MSI-H EC (IA-1 Subset of Patients in IA-2 — Primary Efficacy Analysis).

Kaplan-Meier estimates of OS in the subset of patients from IA-1 with dMMR or MSI-H EC that were included in IA-2. The total number of at-risk patients in the dMMR or MMR-unk/MSI-H EC at 0, 4, 8, 12, 16, 20, 24, 28, 32, and 36 months was 72, 66, 61, 52, 51, 46, 45, 41, 37, 28, 20, 16, 13, 9, 5, 3, and 0, respectively.

Figure 10

KM Plot for OS in Patients With dMMR or MSI-H EC (IA-1 Subset of Patients in IA-2).

Disease Control Rate (IA-1 subset of patients in IA-2)

The results of DCR (based on investigator assessment) for the subset of patients from IA-1 included in IA-2 were 58.3%. In total there were 10 patients with CRs (13.9%), 23 patients with PRs (31.9%), and 9 patients with stable disease (12.5%).

Objective Response Rate (IA-1 subset of patients in IA-2)

The ORR of the subset of patients included from IA-1 (median follow-up time 19.2 months) was 45.8%.

Table 45. Tumour Response Summary in Patients With dMMR or MSI-H EC — RECIST 1.1 Assessed by BICR (IA-1 Subset of Patients in IA-2– Primary Efficacy Analysis).

Table 45

Tumour Response Summary in Patients With dMMR or MSI-H EC — RECIST 1.1 Assessed by BICR (IA-1 Subset of Patients in IA-2– Primary Efficacy Analysis).

PFS (IA-1 Subset of Patients in IA-2)

In the subset of patients from IA-1 (N = 72) included in IA-2 was 52.8% having a PFS event and median PFS of 12.2 months (95% CI, 3.0 to NR).

Table 46. KM Analysis of PFS in Patients With dMMR or MSI-H EC — RECIST 1.1 Based on BICR (IA-1 Subset of Patients in IA-2 – Primary Efficacy Analysis).

Table 46

KM Analysis of PFS in Patients With dMMR or MSI-H EC — RECIST 1.1 Based on BICR (IA-1 Subset of Patients in IA-2 – Primary Efficacy Analysis).

Duration of Response (IA-1 Subset of Patients With an Objective Response in IA-2)

The results of the DOR for the subset of patients from IA-1 included in IA-2 who had an objective response (N = 33) was similar to that of the full cohort. Median DOR was not reached in this subgroup of the population either. With a median follow-up of 19.2 months, 84.8% of the responders had an ongoing response at the time of IA-2. Approximately 91% of patients with a response had a DOR of ≥ 6 months.

Table 47. KM Analysis of DOR in Patients With dMMR or MSI-H EC — RECIST 1.1 Based on BICR (IA-1 Subset of Patients With an Objective Response at IA-2 – Primary Efficacy Analysis).

Table 47

KM Analysis of DOR in Patients With dMMR or MSI-H EC — RECIST 1.1 Based on BICR (IA-1 Subset of Patients With an Objective Response at IA-2 – Primary Efficacy Analysis).

Table 48. Tumour Response Summary in Patients With dMMR or MSI-H EC — irRECIST Based on Investigators’ Assessment (Secondary Efficacy Analysis Set).

Table 48

Tumour Response Summary in Patients With dMMR or MSI-H EC — irRECIST Based on Investigators’ Assessment (Secondary Efficacy Analysis Set).

Table 49. KM Analysis of irDOR in Patients With dMMR or MSI-H EC — irRECIST Based on Investigators’ Assessment (Secondary Efficacy Analysis Set — Patients With Objective Response).

Table 49

KM Analysis of irDOR in Patients With dMMR or MSI-H EC — irRECIST Based on Investigators’ Assessment (Secondary Efficacy Analysis Set — Patients With Objective Response).

Kaplan-Meier estimates of irPFS at IA-2 in patients with dMMR or MSI-H EC based on investigators’ assessment for the secondary efficacy analysis dataset. The total number of at-risk patients in the dMMR or MMR-unk/MSI-H EC at 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, and 36 months was 99, 71, 57, 53, 42, 35, 29, 26,18, 13, 87, 68, 46, 37, 20, 13, 13, 10, 7, 4, 2, 0, 0, and 0, respectively.

Figure 11

KM Plot for irPFS in Patients With dMMR or MSI-H EC — irRECIST Based on Investigators’ Assessment (IA-2, Secondary Efficacy Analysis Set).

Table 50. Sample Size and Baseline Characteristics After Matching — Initial RWE Cohort (Base-Case and Scenario Analysis).

Table 50

Sample Size and Baseline Characteristics After Matching — Initial RWE Cohort (Base-Case and Scenario Analysis).

Table 51. Sample Size and Baseline Characteristics After Matching — Treatment-Specific RWE Cohort.

Table 51

Sample Size and Baseline Characteristics After Matching — Treatment-Specific RWE Cohort.

Table 52. Baseline Characteristics Following Matching for Each MAIC Versus Published Literature.

Table 52

Baseline Characteristics Following Matching for Each MAIC Versus Published Literature.

Table 53. Propensity Score Model (Logistic Regression) — Lean and Full Models GARNET Versus Flatiron Real-World Cohort.

Table 53

Propensity Score Model (Logistic Regression) — Lean and Full Models GARNET Versus Flatiron Real-World Cohort.

Table 54. Baseline and Prognostic Factors Considered for Analyses — GARNET Versus Flatiron Real-World Cohort (After IPTW/PSM [Lean Model]).

Table 54

Baseline and Prognostic Factors Considered for Analyses — GARNET Versus Flatiron Real-World Cohort (After IPTW/PSM [Lean Model]).

Two bar graphs illustrating the distribution of propensity scores of the GARNET cohort versus the Flatiron real-world cohort.

Figure 12

Distribution of Propensity Scores GARNET Versus Flatiron Real-World Cohort (Lean Model).

Two bar graphs illustrating the distribution of propensity scores of the GARNET cohort versus the Flatiron real-world cohort.

Figure 13

Distribution of Propensity Scores GARNET Versus Flatiron Real-World Cohort (Full Model).

Table 55. Regression Models for OS Before and After IPTW/PSM — GARNET Versus Flatiron Cohort.

Table 55

Regression Models for OS Before and After IPTW/PSM — GARNET Versus Flatiron Cohort.

Table 56. Regression Models for OS Adjusting for Covariates — GARNET Versus Flatiron Real-World Cohort.

Table 56

Regression Models for OS Adjusting for Covariates — GARNET Versus Flatiron Real-World Cohort.

Table 57. Regression Models for OS Adjusting for Covariates and Propensity Scores — GARNET Versus Real-World Cohort.

Table 57

Regression Models for OS Adjusting for Covariates and Propensity Scores — GARNET Versus Real-World Cohort.

Table 58. Baseline Characteristics After IPTW — GARNET Versus NCRAS Base-Case Propensity Score Models (ATE).

Table 58

Baseline Characteristics After IPTW — GARNET Versus NCRAS Base-Case Propensity Score Models (ATE).

Appendix 5. Data From Sponsor (IA-3)

Note that this appendix has not been copy-edited.

Table 59. Patient Population EC (A1) Data Cut-Off November 1, 2021.

Table 59

Patient Population EC (A1) Data Cut-Off November 1, 2021.

Table 60. Patient Disposition Safety Analysis Set, Data Cut-Off November 1, 2021.

Table 60

Patient Disposition Safety Analysis Set, Data Cut-Off November 1, 2021.

Table 61. Demographics and Baseline Characteristics EC Safety Analysis Set, Data Cut-Off November 1, 2021.

Table 61

Demographics and Baseline Characteristics EC Safety Analysis Set, Data Cut-Off November 1, 2021.

Table 62. Efficacy — ORR (RECIST 1.1 by BICR) Primary Efficacy Analysis Set, Data Cut-Off November 1, 2021.

Table 62

Efficacy — ORR (RECIST 1.1 by BICR) Primary Efficacy Analysis Set, Data Cut-Off November 1, 2021.

Table 63. Efficacy — Duration of Response (RECIST 1.1 by BICR) Primary Efficacy Analysis Set, Data Cut-Off November 1, 2021.

Table 63

Efficacy — Duration of Response (RECIST 1.1 by BICR) Primary Efficacy Analysis Set, Data Cut-Off November 1, 2021.

Table 64. Overall Summary of TEAEs — Data Cut-Off November 1, 2021(Safety Analysis Set).

Table 64

Overall Summary of TEAEs — Data Cut-Off November 1, 2021(Safety Analysis Set).

Table 65. TEAEs Regardless of Causality — Safety Analysis Set, at Least 15% Patients, Data Cut-Off November 1, 2021.

Table 65

TEAEs Regardless of Causality — Safety Analysis Set, at Least 15% Patients, Data Cut-Off November 1, 2021.

Table 66. Related TEAEs EC (A1) Safety Analysis Set, at Least 5% Patients, Data Cut-Off November 1, 2021.

Table 66

Related TEAEs EC (A1) Safety Analysis Set, at Least 5% Patients, Data Cut-Off November 1, 2021.

Table 67. Grade 3 or Higher TEAEs Regardless of Causality — Safety Analysis Set, at Least 2% Patients, Data Cut-Off November 1, 2021,.

Table 67

Grade 3 or Higher TEAEs Regardless of Causality — Safety Analysis Set, at Least 2% Patients, Data Cut-Off November 1, 2021,.

Table 68. Serious TEAEs Regardless of Causality — Safety Analysis Set, at Least 2% Patients, Data Cut-Off November 1, 2021.

Table 68

Serious TEAEs Regardless of Causality — Safety Analysis Set, at Least 2% Patients, Data Cut-Off November 1, 2021.

Table 69. Summary of Overall irAEs Regardless of Causality — Safety Analysis Set, Over 2 Patients, Data Cut-Off November 1, 2021.

Table 69

Summary of Overall irAEs Regardless of Causality — Safety Analysis Set, Over 2 Patients, Data Cut-Off November 1, 2021.

Table 70. Summary of AEs Leading to Death — Safety Analysis Set Data, Cut-Off November 1, 2021.

Table 70

Summary of AEs Leading to Death — Safety Analysis Set Data, Cut-Off November 1, 2021.

Table 71. Progression-Free Survival (RECIST 1.1 by BICR) Primary Efficacy Analysis Set EC (A1 and A2), Data Cut-Off November 1, 2021.

Table 71

Progression-Free Survival (RECIST 1.1 by BICR) Primary Efficacy Analysis Set EC (A1 and A2), Data Cut-Off November 1, 2021.

Kaplan-Meier estimates of PFS at IA-3 in patients with dMMR or MSI-H EC for the Secondary efficacy analysis dataset.

Figure 14

KM Plot for Progression-Free Survival per RECIST 1.1 by BICR) — Primary Efficacy Analysis Set, Data Cut-Off November 1, 2021.

Table 72. OS (Cohort A1 and A2) — Primary Efficacy Analysis Set, Data Cut-Off November 1, 2021.

Table 72

OS (Cohort A1 and A2) — Primary Efficacy Analysis Set, Data Cut-Off November 1, 2021.

Kaplan-Meier estimates of OS at IA-3 in patients with dMMR or MSI-H EC for the primary efficacy analysis dataset. The total number of at-risk patients in the dMMR or MMR-unk/MSI-H EC at 0, 4, 8, 12, 16, 20, 24, 28, 32, and 36 months was 122, 103, 86, 75, 62, 54, 46, 35, 29, 19, 10, 4, 0, and 0, respectively.

Figure 15

Kaplan-Meier Survival Plot for OS — Primary Efficacy Analysis Set, Data Cut-Off November 1, 2021.

References

1.
Plaxe S, Mundt A. Overview of endometrial carcinoma. In: Post TW, ed. UptoDate. Waltham (MA): UptoDate; 2021: www​.uptodate.com. Accessed 2021 Dec 16.
2.
Canadian Cancer Society. Uterine cancer statistics. 2021; https://cancer​.ca/en​/cancer-information/cancer-types​/uterine/statistics. Accessed 2021 Dec 17.
3.
4.
Fung-Kee-Fung M, Dodge J, Elit L, et al. Follow-up after primary therapy for endometrial cancer: a systematic review. Gynecol Oncol. 2006;101(3):520-529. [PubMed: 16556457]
5.
Endometrial cancer. Edmonton (AB): Alberta Health Services; 2015: https://www​.albertahealthservices​.ca/assets​/info/hp/cancer/if-hp-cancer-guide-gyne002-endometrial​.pdf. Accessed 2022 Jan 14.
6.
Canadian Cancer Statistics Advisory Committee. Canadian cancer statistics: a 2018 special report on cancer indicence by stage. Toronto (ON): Canadian Cancer Society, Statistics Canada, the Public Health Agency of Canada; 2018: https://cdn​.cancer.ca​/-/media/files/research​/cancer-statistics​/2018-statistics/canadian-cancer-statistics-2018-en.pdf. Accessed 2021 Dec 30.
7.
Green AK, Feinberg J, Makker V. A Review of Immune Checkpoint Blockade Therapy in Endometrial Cancer. Am Soc Clin Oncol Educ Book. 2020;40:1-7. [PubMed: 32213091]
8.
Lorenzi M, Amonkar M, Zhang J, Mehta S, Liaw K-L. Epidemiology of Microsatellite Instability High (MSI-H) and Deficient Mismatch Repair (dMMR) in Solid Tumors: A Structured Literature Review. J Oncol. 2020;2020:1807929.
9.
Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J Hematol Oncol. 2019;12(1):54. [PMC free article: PMC6544911] [PubMed: 31151482]
10.
Sovak MA, Dupont J, Hensley ML, et al. Paclitaxel and carboplatin in the treatment of advanced or recurrent endometrial cancer: a large retrospective study. Int J Gynecol Cancer. 2007;17(1):197-203. [PubMed: 17291253]
11.
Sorbe B, Andersson H, Boman K, Rosenberg P, Kalling M. Treatment of primary advanced and recurrent endometrial carcinoma with a combination of carboplatin and paclitaxel-long-term follow-up. Int J Gynecol Cancer. 2008;18(4):803-808. [PubMed: 17944917]
12.
Makker V, Hensley ML, Zhou Q, Iasonos A, Aghajanian CA. Treatment of advanced or recurrent endometrial carcinoma with doxorubicin in patients progressing after paclitaxel/carboplatin: Memorial Sloan-Kettering Cancer Center experience from 1995 to 2009. Int J Gynecol Cancer. 2013;23(5):929-934. [PMC free article: PMC3694716] [PubMed: 23598889]
13.
Miller DS, Filiaci VL, Mannel RS, et al. Carboplatin and Paclitaxel for Advanced Endometrial Cancer: Final Overall Survival and Adverse Event Analysis of a Phase III Trial (NRG Oncology/GOG0209). J Clin Oncol. 2020;38(33):3841-3850. [PMC free article: PMC7676887] [PubMed: 33078978]
14.
Brooks RA, Fleming GF, Lastra RR, et al. Current recommendations and recent progress in endometrial cancer. CA Cancer J Clin. 2019;69(4):258-279. [PubMed: 31074865]
15.
Jemperli (dostarlimab): solution for infusion, 500 mg/10 mL vial (50 mg/mL) [product monograph]. Mississauga (ON): GlaxoSmithKline Inc.; 2021 Mar 26.
16.
National Institute for Health Care and Excellence. Dostarlimab for previously treated advanced or recurrent endometrial cancer with high microsatellite instability or mismatch repair deficiency [ID3802]. 2021; https://www​.nice.org​.uk/guidance/indevelopment/gid-ta10670. Accessed 2021 Dec 20.
17.
Dostarlimab (Jemperli). Glasgow (GB): Scottish Medicines Consortium; 2021: https://www​.scottishmedicines​.org.uk/medicines-advice​/dostarlimab-jemperli-full-smc2404/ Accessed 2021 Dec 20.
18.
Clinical Study Report: 4010-01-001. A phase 1 dose escalation and cohort expansion study OF TSR-042, an anti-PD-1 monoclonal antibody, in patients with advanced solid tumors - part 2B, endometrial cancer (cohorts A1 and A2) [internal sponsor's report]. Barcelona (ES): GlaxoSmithKline Research & Development Limited; 2020 Nov 16.
19.
Sponsor's ITC reports [internal sponsor's report]. In: Jemperli, solution for infusion, 500 mg/10 mL vial (50 mg/mL) [internal sponsor's package]. Mississauga (ON): GlaxoSmithKline Inc; 2021 Sep 22.
20.
Canadian Cancer Society. Cancerous tumours of the uterus. 2021; https://cancer​.ca/en​/cancer-information/cancer-types​/uterine​/what-is-uterine-cancer​/cancerous-tumours. Accessed 2021 Dec 17.
21.
American Society of Clinical Oncology. Uterine cancer: risk factors and prevention. 2020; https://www​.cancer.net​/cancer-types/uterine-cancer​/risk-factors-and-prevention. Accessed 2021 Dec 20.
22.
American Cancer Society. Endometrial cancer stages. 2019; https://www​.cancer.org​/cancer/endometrial-cancer​/detection-diagnosis-staging​/staging.html Accessed 2021 Dec 22.
23.
Canadian Cancer Society. Stages of uterine cancer. 2021; https://cancer​.ca/en​/cancer-information/cancer-types​/uterine/staging. Accessed 2021 Dec 22.
24.
American Cancer Society. Endometrial cancer: detection, diagnosis, staging, survival rates. 2021; https://www​.cancer.org​/cancer/endometrial-cancer​/detection-diagnosis-staging​/survival-rates.html1. Accessed 2021 Dec 17.
25.
McEachron J, Zhou N, Spencer C, et al. Adjuvant chemoradiation associated with improved outcomes in patients with microsatellite instability-high advanced endometrial carcinoma. Int J Gynecol Cancer. 2021;31(2):203-208. [PubMed: 32817172]
26.
Sorbe B, Juresta C, Ahlin C. Natural history of recurrences in endometrial carcinoma. Oncol Lett. 2014;8(4):1800-1806. [PMC free article: PMC4156268] [PubMed: 25202413]
27.
Guidelines & Protocols Advisory Committee. Genital tract cancers in females: endometrial cancer. Victoria (BC): BCGuidelines.ca; 2014: https://www2​.gov.bc.ca​/gov/content/health​/practitioner-professional-resources​/bc-guidelines​/endometrial-cancer. Accessed 2022 Jan 14.
28.
PDQ Adult Treatment Editorial Board. PDQ endometrial cancer treatment. Bethesda (MD): National Cancer Institute; 2021: https://www​.cancer.gov​/types/uterine/hp/endometrial-treatment-pdq#_36 Accessed 2021 Dec 20.
29.
Systemic therapy for advanced or recurrent endometrial cancer and advanced or recurrent uterine papillary serous carcinoma. Toronto (ON): Cancer Care Ontario; 2019: https://www​.cancercareontario​.ca/en/guidelines-advice​/types-of-cancer/501. Accessed 2022 Jan 14.
30.
Fleming GF, Brunetto VL, Cella D, et al. Phase III trial of doxorubicin plus cisplatin with or without paclitaxel plus filgrastim in advanced endometrial carcinoma: a Gynecologic Oncology Group Study. J Clin Oncol. 2004;22(11):2159-2166. [PubMed: 15169803]
31.
Thigpen JT, Brady MF, Homesley HD, et al. Phase III trial of doxorubicin with or without cisplatin in advanced endometrial carcinoma: a gynecologic oncology group study. J Clin Oncol. 2004;22(19):3902-3908. [PubMed: 15459211]
32.
van Wijk FH, Aapro MS, Bolis G, et al. Doxorubicin versus doxorubicin and cisplatin in endometrial carcinoma: definitive results of a randomised study (55872) by the EORTC Gynaecological Cancer Group. Ann Oncol. 2003;14(3):441-448. [PubMed: 12598351]
33.
Concin N, Matias-Guiu X, Vergote I, et al. ESGO/ESTRO/ESP guidelines for the management of patients with endometrial carcinoma. Int J Gynecol Cancer. 2021;31(1):12-39. [PubMed: 33397713]
34.
Center for Drug Evaluation and Research. Multi-discipline review(s). Jemperli (dostarlimab-gxly). Company: GlaxoSmithKline LLC. Application No. 761223. Approval date: 17/08/2021 (FDA drug approval package). Rockville (MD): U.S. Food and Drug Administration (FDA); 2021: https://www​.accessdata​.fda.gov/scripts/cder/daf/index​.cfm?event=overview​.process&varApplNo​=761223. Accessed 2021 Dec 20.
35.
European Public Assessment Report (EPAR): Jemperli (dostarlimab). London (GB): European Medicines Agency; 2021: https://www​.ema.europa​.eu/en/medicines/human/EPAR/jemperli Accessed 2021 Dec 20.
36.
Carboplatin injection BP (carboplatin injection): 10 mg / mL (50 mg, 150 mg, 450 mg, 600 mg of carboplatin per vial) [product monograph]. Kirkland (QC): Pfizer Canada ULC; 2019: https://www​.pfizer.ca​/sites/default/files​/202001/Carboplatin​_PM_E_231317_16Dec2019.pdf. Accessed 2022 Jan 20.
37.
Paclitaxel injection USP: solution for injection, 6 mg/mL [product monograph]. Kirkland (QC): Accord Healthcare Inc; 2020: https://pdf​.hres.ca/dpd_pm/00057982.PDF. Accessed 2022 Jan 20.
38.
Provera (medroxyprogesterone acetate tablets USP): 2.5 mg, 5 mg and 10 mg tablets [product monograph]. Kirkland (QC): Pfizer Canada Inc; 2014: https://www​.pfizer.ca​/sites/default/files​/201710/Provera_PM_E​_176877_29September2014.pdf. Accessed 2021 Dec 22.
39.
Seymour L, Bogaerts J, Perrone A, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18(3):e143-e152. [PMC free article: PMC5648544] [PubMed: 28271869]
40.
Common terminology criteria for adverse events (CTCAE): version 5.0. Rockville (MD): U.S. Department of Health and Human Services, National Institutes of Health, National Cancer Institute; 2017: https://ctep​.cancer.gov​/protocoldevelopment​/electronic_applications​/docs/CTCAE_v5_Quick_Reference_8​.5x11.pdf. Accessed 2022 Jan 14.
41.
Oken MM, Creech RH, Tormey DC, et al. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 1982;5(6):649-655. [PubMed: 7165009]
42.
McGowan J, Sampson M, Salzwedel DM, Cogo E, Foerster V, Lefebvre C. PRESS Peer Review of Electronic Search Strategies: 2015 guideline statement. J Clin Epidemiol. 2016;75:40-46. [PubMed: 27005575]
43.
Grey matters: a practical tool for searching health-related grey literature. Ottawa (ON): CADTH; 2019: https://www​.cadth.ca/grey-matters. Accessed 2022 Jan 14.
44.
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]
45.
Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228-247. [PubMed: 19097774]
46.
Drug Reimbursement Review sponsor submission: pemigatinib, 4.5 mg, 9 mg, and 13.5 mg, oral tablets [internal sponsor's package]. Pointe Claire (QC): Incyte Biosciences Canada Corporation; 2021 Jun 23.
47.
Canadian Cancer Trial Group. irRECIST. 2021; https://recist​.eortc.org/irecist/. Accessed 2022 Jan 17.
48.
EORTC quality of life. https://qol​.eortc.org/. Accessed 2021 Nov 22.
49.
Brooks R. EuroQol: the current state of play. Health Policy. 1996;37(1):53-72. [PubMed: 10158943]
50.
Clinical Study Report: 4010-01-001. A phase 1 dose escalation and cohort expansion study of TSR-042, an anti-PD-1 monoclonal antibody, in patients with advanced solid tumors - part 2B, non-endometrial cancer (cohort F) [internal sponsor's report]. Paris (FR): GlaxoSmithKline Research & Development Limited; 2020 Nov 13.
51.
Cochrane Handbook for Systematic Reviews of Interventions. 2nd Edition. In: Higgins JPT, Thomas J, Chandler J, et al., eds. Chichester (UK): John Wiley & Sons; 2019: https://training​.cochrane.org/handbook. Accessed 2022 Jan 6.
52.
National Institute for Health and Care Excellence. The guidelines manual: process and methods [PMG6]. 2012: https://www​.nice.org​.uk/process/pmg6/chapter/introduction. Accessed 2022 Jan 6.
53.
Programme CAS. CASP Cohort Study Checklist. 2018: https://casp-uk​.net/wp-content​/uploads/2018​/01/CASP-Cohort-Study-Checklist_2018​.pdf. Accessed 2022 Jan 6.
54.
Signorovitch JE, Wu EQ, Yu AP, et al. Comparative effectiveness without head-to-head trials: a method for matching-adjusted indirect comparisons applied to psoriasis treatment with adalimumab or etanercept. Pharmacoeconomics. 2010;28(10):935-945. [PubMed: 20831302]
55.
Guyot P, Ades AE, Ouwens MJNM, Welton NJ. Enhanced secondary analysis of survival data: reconstructing the data from published Kaplan-Meier survival curves. BMC Med Res Methodol. 2012;12(1):9. [PMC free article: PMC3313891] [PubMed: 22297116]
56.
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]
57.
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]
58.
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]
59.
Mortimer KM, Neugebauer R, van der Laan M, Tager IB. An application of model-fitting procedures for marginal structural models. Am J Epidemiol. 2005;162(4):382-388. [PubMed: 16014771]
60.
Brumback BA, Hernán MA, Haneuse SJ, Robins JM. Sensitivity analyses for unmeasured confounding assuming a marginal structural model for repeated measures. Stat Med. 2004;23(5):749-767. [PubMed: 14981673]
61.
Phillippo DM, Ades AE, Dias S, Palmer S, Abrams KR, Welton NJ. NICE DSU technical support document 18: methods for population-adjusted indirect comparisons in submissions to NICE. Sheffield (GB): NICE Decision Support Unit; 2016: http://nicedsu​.org.uk​/wp-content/uploads​/2017/05/Population-adjustment-TSD-FINAL.pdf. Accessed 2022 Jan 7.
62.
Aaronson NK, Ahmedzai S, Bergman B, et al. The European Organization for Research and Treatment of Cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in oncology. J Natl Cancer Inst. 1993;85(5):365-376. [PubMed: 8433390]
63.
Davda J, Kibet H, Achieng E, Atundo L, Komen T. Assessing the acceptability, reliability, and validity of the EORTC Quality of Life Questionnaire (QLQ-C30) in Kenyan cancer patients: a cross-sectional study. J Patient Rep Outcomes. 2021;5(1):4. [PMC free article: PMC7790948] [PubMed: 33415528]
64.
Osoba D, Zee B, Pater J, Warr D, Kaizer L, Latreille J. Psychometric properties and responsiveness of the EORTC quality of Life Questionnaire (QLQ-C30) in patients with breast, ovarian and lung cancer. Qual Life Res. 1994;3(5):353-364. [PubMed: 7841968]
65.
Butler L, Bacon M, Carey M, Zee B, Tu D, Bezjak A. Determining the relationship between toxicity and quality of life in an ovarian cancer chemotherapy clinical trial. J Clin Oncol. 2004;22(12):2461-2468. [PubMed: 15197209]
66.
Zhao H, Kanda K. Translation and validation of the standard Chinese version of the EORTC QLQ-C30. Qual Life Res. 2000;9(2):129-137. [PubMed: 10983477]
67.
Osoba D, Rodrigues G, Myles J, Zee B, Pater J. Interpreting the significance of changes in health-related quality-of-life scores. J Clin Oncol. 1998;16(1):139-144. [PubMed: 9440735]
68.
Bedard G, Zeng L, Zhang L, et al. Minimal important differences in the EORTC QLQ-C30 in patients with advanced cancer. Asia Pac J Clin Oncol. 2014;10(2):109-117. [PubMed: 23551530]
69.
EuroQol--a new facility for the measurement of health-related quality of life. Health Policy. 1990;16(3):199-208. [PubMed: 10109801]
70.
Ameri H, Yousefi M, Yaseri M, Nahvijou A, Arab M, Akbari Sari A. Mapping the cancer-specific QLQ-C30 onto the generic EQ-5D-5L and SF-6D in colorectal cancer patients. Expert Rev Pharmacoecon Outcomes Res. 2019;19(1):89-96. [PubMed: 30173585]
71.
Sinnott PL, Joyce VR, Barnett PG. Guidebook: preference measurement in economic analysis. Menlo Park (CA): Health Economics Research Center; 2007: https://www​.herc.research​.va.gov/files/BOOK_419.pdf. Accessed 2021 Nov 23.
72.
Lang HC, Chuang L, Shun SC, Hsieh CL, Lan CF. Validation of EQ-5D in patients with cervical cancer in Taiwan. Support Care Cancer. 2010;18(10):1279-1286. [PubMed: 19941008]
73.
Setiawan D, Dusafitri A, Galistiani GF, van Asselt ADI, Postma MJ. Health-Related Quality of Life of Patients with HPV-Related Cancers in Indonesia. Value Health Reg Issues. 2018;15:63-69. [PubMed: 29474181]
74.
Hu X, Jing M, Zhang M, Yang P, Yan X. Responsiveness and minimal clinically important difference of the EQ-5D-5L in cervical intraepithelial neoplasia: a longitudinal study. Health Qual Life Outcomes. 2020;18(1):324. [PMC free article: PMC7531135] [PubMed: 33008423]
75.
Bjordal K, de Graeff A, Fayers PM, et al. A 12 country field study of the EORTC QLQ-C30 (version 3.0) and the head and neck cancer specific module (EORTC QLQ-H&N35) in head and neck patients. EORTC Quality of Life Group. Eur J Cancer. 2000;36(14):1796-1807. [PubMed: 10974628]
76.
EORTC QLC-C30 scoring manual. European Organisation for Research and Treatment of Cancer (EORTC) 2001; https://www​.eortc.org​/app/uploads/sites/2/2018/02/SCmanual​.pdf. Accessed 2021 Nov 23.
77.
Bergman B, Sullivan M, Sörenson S. Quality of life during chemotherapy for small cell lung cancer. II. A longitudinal study of the EORTC Core Quality of Life Questionnaire and comparison with the Sickness Impact Profile. Acta Oncol. 1992;31(1):19-28. [PubMed: 1316770]
78.
Bjordal K, Kaasa S. Psychometric validation of the EORTC Core Quality of Life Questionnaire, 30-item version and a diagnosis-specific module for head and neck cancer patients. Acta Oncol. 1992;31(3):311-321. [PubMed: 1622651]
79.
Greimel E, Nordin A, Lanceley A, et al. Psychometric validation of the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Endometrial Cancer Module (EORTC QLQ-EN24). Eur J Cancer. 2011;47(2):183-190. [PubMed: 20851597]
80.
Gallardo-Rincon D, Toledo-Leyva A, Bahena-Gonzalez A, et al. Validation of the QLQ-EN24 instrument for the assessment of health-related quality of life for women with endometrial cancer in Mexico. Arch Gynecol Obstet. 2021;304(3):773-782. [PubMed: 33582853]
81.
McClure NS, Sayah FA, Xie F, Luo N, Johnson JA. Instrument-Defined Estimates of the Minimally Important Difference for EQ-5D-5L Index Scores. Value Health. 2017;20(4):644-650. [PubMed: 28408007]
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: NBK602691

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...