CADTH Reimbursement Review

Pembrolizumab (Keytruda)

Sponsor: Merck Canada Inc.

Therapeutic area: Classical Hodgkin lymphoma

This multi-part report includes:

Clinical Review

Pharmacoeconomic Review

Clinical Review

Abbreviations

AE

adverse event

ASCT

autologous stem cell transplant

BV

brentuximab vedotin

cHL

classical Hodgkin lymphoma

CI

confidence interval

DOR

duration of response

ECOG

Eastern Cooperative Oncology Group

EORTC QLQ-C30

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

EQ-5D-3L

EuroQol 5-Dimensions 3-Levels questionnaire

ES

effect size

HL

Hodgkin lymphoma

HRQoL

health-related quality of life

IWG

International Working Group

OH-CCO DAC

Ontario Health Hematology Disease Site Drug Advisory Committee (Cancer Care Ontario)

ORR

objective response rate

OS

overall survival

PET

positron emission tomography

PFS

progression-free survival

POGO

Pediatric Oncology Group of Ontario

Executive Summary

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

Table 1: Submitted for Review

Item

Description

Drug product

Pembrolizumab (Keytruda) 200 mg in adults or 2 mg/kg in pediatrics administered intravenously every 3 weeks

Indication

Treatment of adult and pediatric patients with refractory or relapsed cHL, as monotherapy, who have failed ASCT, or who are not candidates for multi-agent salvage chemotherapy and ASCT

Reimbursement request

As above

Health Canada approval status

NOC

Health Canada review pathway

Other expedited pathway: Project Orbis

NOC date

February 5, 2021

Sponsor

Merck Canada

ASCT = autologous stem cell transplant; cHL = classical Hodgkin lymphoma; NOC = Notice of Compliance.

Introduction

The purpose of this report is to summarize the evidence regarding the use of pembrolizumab as monotherapy in adult and pediatric patients with refractory or relapsed classical Hodgkin lymphoma (cHL) who have failed autologous stem cell transplant (ASCT) or who are not candidates for salvage chemotherapy and ASCT. Pembrolizumab is an immune checkpoint inhibitor dosed at 200 mg every 3 weeks in adults and 2 mg/kg every 3 weeks in pediatrics.

The term Hodgkin lymphoma (HL) refers to a group of lymphoid proliferations that share clinical and morphological features that distinguish them from other types of lymphoma. It is estimated that in 2020, 1,000 Canadians were diagnosed with cHL and 100 died from the disease.1 Clinically, HL presents most commonly with enlarged cervical lymph nodes, and spread is generally between contiguous nodal areas. Mediastinal masses and B symptoms (fever, weight loss, and night sweats) are common. A bimodal age distribution is appreciated for HL, with most patients diagnosed between the ages of 15 years and 39 years. A second peak is seen in individuals over the age of 70 years.2 HL is diagnosed by biopsy of an affected tissue or organ. On histopathology, large, atypical, and malignant cells, termed Reed-Sternberg or Hodgkin cells, are observed in a heterogeneous background consisting of non-neoplastic inflammatory cells.3 Subclassification of HL into cHL (nodular sclerosis, mixed cellularity, lymphocyte rich, and lymphocyte depleted) and nodular lymphocyte-predominant HL is based on the degree of atypia of the malignant cells, their immunophenotype, and the features of the inflammatory background.

Standards of Therapy

The treatment of cHL is guided by careful assessment of stage- and disease-specific risk factors. 18F-fluorodeoxyglucose PET is considered the gold standard for staging of HL and the Cotswold modification of the Ann Arbor staging system is applied to determine the numerical stage. In 1998, Hasenclever et al. published a prognostic score for adult patients with advanced HL consisting of 7 clinical (sex, age, and stage) and laboratory (anemia, leukocytosis, reduced serum albumin, and lymphopenia) factors. This score predicts freedom from progression (84% for patients with no risk factors to 42% for those with 5 or more factors) and overall survival (OS; 89% for patients with no risk factors to 56% for those with 5 or more factors) at 5 years.4 Treatment is risk-adapted, with low-risk patients (stage I to II, few risk factors) receiving a limited number of cycles of ABVD (doxorubicin-bleomycin-vinblastine-dacarbazine) chemotherapy, often with low-dose involved field radiotherapy5,6 while those with higher-risk disease receiving more extensive chemotherapy. Intensive chemotherapy, in the form of escalated BEACOPP (bleomycin-etoposide-doxorubicin-cyclophosphamide-vincristine-procarbazine-prednisone), is reserved for patients with the highest risk of adverse outcome.7 Response-adapted therapy with the use of interim PET restaging is used to de-escalate treatment for patients who are likely cured8,9 and to escalate treatment of patients who are not responding as expected.8,10 Risk- and response-adapted combined modality therapies are similarly used in pediatric protocols with some differences in chemotherapy backbones.11 Recognizing the shared clinical-pathological features of HL in the most affected age groups, North American adult and pediatric study groups have amalgamated efforts to study new agents using a common chemotherapy backbone.12 The outcome of HL has improved significantly over time and today more than 80% of patients with cHL are cured with initial therapy.2,13

Most patients who do not respond to or who relapse after first-line treatment for HL are treated with high-dose chemotherapy and autologous cell transplantation.14 This approach is supported by 2 randomized trials,15,16 several phase II17-20 and registry21 studies, and results in progression-free survival (PFS) in 50% to 60% of patients and OS in 60% to 80% of patients. Careful patient selection is required for successful ASCT as the presence of multiple or severe comorbidities may make the treatment-related mortality of high-dose therapy prohibitive. Patients may also not undergo ASCT if they fail to mobilize sufficient numbers of hematopoietic stem cells to support the use of high-dose chemotherapy, if they fail to respond to salvage chemotherapy, or for reasons of conscience as in the case of Jehovah’s Witnesses. Overall, approximately 85% of patients with relapsed or refractory cHL undergo ASCT.

There is currently no standard of care for patients with cHL who relapse after ASCT or who are ineligible for ASCT for 1 of the reasons noted above. Options to treat these patients include chemotherapy, radiotherapy (for those with localized recurrences), targeted therapy with brentuximab vedotin (BV), and immune checkpoint inhibitors. Although responses to standard-dose chemotherapy occur frequently in later lines of treatment, the use of conventional-dose salvage chemotherapy is unlikely to lead to a cure in these patients.22 Good palliation can be achieved with oral single-agent or combination chemotherapy regimens.23,24 BV is an antibody-drug conjugate that targets CD30-positive cells, delivering the antimitotic agent MMAE into the cytoplasm of these cells by endocytosis.25 The utility of BV in relapsed or refractory cHL was demonstrated in a pivotal phase II study in 102 patients who failed ASCT. The overall response rate was 75% with complete responses seen in 34% of patients and toxicity was manageable, although peripheral neuropathy was frequently dose limiting.26 After 5 years of follow-up, OS in this cohort of 102 patients was 41% and PFS was 22%; median OS and PFS were higher among those patients who achieved complete remission to BV.27 Real-world experience with BV in transplant-ineligible relapsed or refractory cHL was provided in a phase II study of 136 patients with a median age of 70 years at diagnosis. The most common reasons for transplant ineligibility in this cohort were comorbidities and age. A median of 8 cycles was given and overall and complete responses were observed in 74.3% and 34.6% of patients, respectively, similar to the results seen in patients who had previously undergone ASCT. Median PFS and OS were 15.1 and 17.8 months, respectively.28

Immune checkpoint inhibitors affect the PD1/PD-L1 axis and lead to increased immune reactivity against cancers that have exploited this mechanism to escape immune control. Nivolumab is a human IgG monoclonal antibody that targets PD1. Nivolumab is licensed for treatment of patients with advanced hepatocellular carcinoma, non–small cell lung cancer, advanced renal cell carcinoma, and certain cases of colorectal carcinoma or malignant melanoma. It is also approved for treatment of patients with cHL who have progressed after ASCT and BV or 3 or more lines of systemic therapy including hematopoietic cell transplantation.29 The indication in cHL is based on the results of the CHECKMATE-205 and CHECKMATE-039 studies, which enrolled a total of 95 patients, demonstrating an overall response rate of 66% and complete and partial remission of 6% and 60%, respectively. Median duration of response (DOR) was 13.1 months. Toxicity was manageable, although immune-mediated toxicity was observed.30,31 A second immune checkpoint inhibitor, pembrolizumab, is the subject of this CADTH review.

Stakeholder Perspectives

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

Patient Input

Lymphoma Canada was the only patient group to provide input and did so after conducting 2 online surveys which yielded 128 responses. Patients often experienced fatigue, trouble breathing, fever/chills, loss of appetite, itching, anxiety, problems concentrating, loss of sexual desire, and memory loss. Many patients had quit working or school due to their diagnosis. Patients sought treatments that would provide disease control or remission with fewer side effects than current treatment options and valued longer survival and remission.

Clinician Input

Input From Clinical Experts Consulted by CADTH

The clinical experts highlighted that patients with cHL who relapse after ASCT or who are not fit for multi-agent chemotherapy and ASCT have limited treatment options. The available treatments can be associated with significant side effects and are seldom curative. Both pediatric and adult experts expect pembrolizumab to be effective earlier in the treatment paradigm but that it is also appropriate for use in patients who have failed or are ineligible for ASCT. However, patients recently on therapy for autoimmune disease, patients with poor performance status, patients with organ failure, or at high risk of autoimmune side effects may not be suited for pembrolizumab. A response to therapy would be marked by resolution of disease symptoms, radiologic evidence of disease improvement, improved ability to perform activities of daily living, reduction in size of lymph nodes and other disease sites, and in some patients, becoming eligible for an allogeneic or ASCT. Patients receiving pembrolizumab are assessed clinically every 3 weeks and radiologically every 3 to 4 cycles. Treatment should be discontinued if there is disease progression, severe immune-related adverse event (AE), or severe infusion or hypersensitivity reactions.

Clinician Group Input

Twelve clinicians from the Ontario Health Hematology Disease Site Drug Advisory Committee, Lymphoma Canada Scientific Advisory Board, and the Pediatric Oncology Group of Ontario (POGO) all provided feedback for this review. The input provided aligned with the advice provided from the CADTH clinical experts.

Drug Program Input

Some drug plan questions were regarding retreating patients with pembrolizumab who had already received it. The clinical experts identified limited evidence to provide guidance, but there is some evidence of retreating patients with pembrolizumab who have already received 35 cycles if disease progression is observed. The clinical experts were hesitant to treat patients with pembrolizumab if they had already been treated with a PD-1 or PD-L1 inhibitor unsuccessfully, believing pembrolizumab should be stopped if there is evidence of disease progression or intolerable side effects. The clinical experts were also hesitant to switch a patient from BV to pembrolizumab if the patient is responding to BV.

Clinical Evidence

Description of Studies

KEYNOTE-051

The KEYNOTE-05132 study was a nonrandomized, open-label, single-arm trial of pembrolizumab 2 mg/kg administered every 3 weeks in 7 pediatric patients aged 3 years to 18 years with relapsed or refractory cHL. A 28-day screening period was performed before patient enrolment to collect necessary laboratory, diagnostic, and demographic information and assess study eligibility. The KEYNOTE-051 study evaluated safety and efficacy including objective response rate (ORR), DOR, PFS, and OS for 35 cycles of treatment or until discontinuation due to disease progression or AEs. Post-treatment follow-up assessments occurred every 12 weeks. The study was funded by the sponsor and had a data cut-off date of January 2020.

KEYNOTE-087

The KEYNOTE-08733 study was a nonrandomized, single-arm study of pembrolizumab 200 mg administered every 3 weeks in adult patients with cHL. A 28-day screening period was performed before patient enrolment to collect necessary laboratory, diagnostic, and demographic information and assess study eligibility. The study evaluated ORR, PFS, DOR, health-related quality of life (HRQoL), and OS with a treatment duration up to 2 years, or until discontinuation of treatment due to disease progression, or occurrence of AEs. Post-treatment follow-up assessments occurred every 12 weeks. The study was funded by the sponsor with a data cut-off date of March 2019. The study consisted of 3 cohorts:

KEYNOTE-204

The KEYNOTE-20434 study was a phase III, randomized (1:1 ratio), active controlled, open-label clinical trial comparing pembrolizumab 200 mg administered intravenously every 3 weeks (N = 151) with BV 1.8 mg/kg (maximum dose of 180 mg) administered intravenously every 3 weeks (N = 153) in adult patients with relapsed or refractory cHL. A 28-day screening period was performed before patient enrolment to collect necessary laboratory, diagnostic, and demographic information and assess study eligibility. The study evaluated PFS, OS, ORR, DOR, time to response, HRQoL, and safety for 35 cycles of treatment or until early discontinuation due to disease progression, unacceptable AEs, or other reasons to withdraw therapy. Post-treatment follow-up assessments occurred every 12 weeks. The study was funded by the sponsor with data cut-off date of February 2020. A diagram of the KEYNOTE-204 study design is provided in Figure 2.

Baseline Characteristics

Patients in the KEYNOTE-051 study had a median age of 15 years while the median age in the KEYNOTE-087 and KEYNOTE-204 studies ranged from 32.0 to 40.0. The proportion of female patients ranged from 41.2% among BV patients in the KEYNOTE-204 study to 47.8% among cohort 1 of the KEYNOTE-087 study. The proportion of patients with an Eastern Cooperative Oncology Group (ECOG) score of 0 ranged from 42.0% in cohort 1 of the KEYNOTE-087 study to 65.4% among BV patients from the KEYNOTE-204 study. The proportion of patients with an ECOG score of 0 was 54.3% and 48.3% in cohorts 2 and 3, respectively, of the KEYNOTE-087 study and 57.0% in the pembrolizumab arm of the KEYNOTE-204 study. Cohorts 1 and 3 of the KEYNOTE-087 study had higher rates of prior radiation use (46.4% and 40.0%, respectively) relative to either arm in the KEYNOTE-204 study (pembrolizumab: 38.4% and BV: 39.9%) while those in cohort 2 had lower rates (25.9%). Patients in either arm of the KEYNOTE-204 study had more bulky disease (pembrolizumab: 23.2% and BV: 16.3%) relative to any cohort in the KEYNOTE-087 study (cohort 1: 2.9%, cohort 2: 6.2%, and cohort 3: 1.7%). Baseline B symptoms were present in 30.4%, 33.3%, and 31.7% of patients in cohort 1, cohort 2, and cohort 3 of the KEYNOTE-087 study. Baseline B symptoms were also present in 28.5% and 23.5% of pembrolizumab and BV patients, respectively, in the KEYNOTE-204 study. The 2 arms within the KEYNOTE-204 study seem relatively balanced except that pembrolizumab patients had higher rates of bulky disease (23.2% versus 16.3%). Patients in the KEYNOTE-204 study were permitted to be treated with a subsequent anticancer medication after pembrolizumab or BV was discontinued.

Efficacy Results

Progression-Free Survival

In KEYNOTE-051, 3 patients (42.9%) experienced an event (disease progression or death). In the KEYNOTE-087 study, there were 43 (62.3%), 54 (66.7%), and 36 (60.0%) events in cohorts 1, 2, and 3, respectively. In the KEYNOTE-204 study, the proportion of patients experiencing an event was similar between the pembrolizumab (53.6%) and BV (57.5%) arms. In the KEYNOTE-051 study, the median PFS was reported to be 11.1 months (95% confidence interval [CI], 2.6 to not reported). In the KEYNOTE-087 study, median survival was reported to be 16.4 months (95% CI, 11.3 to 27.6), 11.1 months (95% CI, 7.3 to 13.5), and 19.4 (95% CI, 8.4 to 22.1) months in cohorts 1, 2, and 3, respectively. In the KEYNOTE-204 study, the median PFS was higher in the pembrolizumab arm (13.2 months; 95% CI, 10.9 to 19.4) than the BV arm (8.3 months; 95% CI, 5.7 to 8.8). In the KEYNOTE-051 study, the PFS rate at 12 months was 27.8% (no 95% CI reported). In the KEYNOTE-087 study, the PFS rate at 12 months was 61.3%, 43.0%, and 53.9% in cohorts 1, 2, and 3, respectively (no 95% CI reported). In the KEYNOTE-204 study, the 12-month PFS rate was higher in the pembrolizumab arm (53.9%; 95% CI, 45.0 to 61.9) than the BV arm (35.6%; 95% CI, 26.9 to 44.4). In the KEYNOTE-087 study, the 24-month PFS rate was 41.6%, 21.9%, and 34.0% in cohorts 1, 2, and 3, respectively (no 95% CI reported). In the KEYNOTE-204 study, the 24-month PFS rate was 35.4% (95% CI, 26.2 to 44.6) in the pembrolizumab arm and 25.4% (95% CI, 17.1 to 34.5) in the BV arm. The hazard ratio for time to progression was 0.65 (95% CI, 0.48 to 0.88), which was statistically significant (P = 0.0027).

Overall Survival

In the KEYNOTE-051 study, minimal information regarding OS was provided. In the KEYNOTE-087 study, 15.9%, 16.0%, and 15.0% of patients in cohorts 1, 2, and 3, respectively, died. In the KEYNOTE-204 study, a smaller proportion of patients receiving pembrolizumab died relative to patients receiving BV (10.6% versus 19.6%). Median survival was not reported in the KEYNOTE-051 study and not reached in the KEYNOTE-087 or KEYNOTE-204 studies. In the KEYNOTE-051 study, 100% of patients were alive at 12 months. In the KEYNOTE-087 study, OS at 12 months was 95.7%, 96.,2% and 96.6% in cohorts 1, 2, and 3, respectively (95% CI not reported). |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| At 24 months in the KEYNOTE-087 study, 92.6%, 91.0%, and 89.4% of patients were alive in cohorts 1, 2, and 3, respectively (95% CI not reported).  |  |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||| |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||

Objective Response Rate

In the KEYNOTE-051 study, 42.9% (95% CI, 9.9 to 81.6) of patients experienced a partial or complete response. In the KEYNOTE-087 study, 78.3% (95% CI, 66.7 to 87.3), 64.2% (95% CI, 52.8 to 74.6), and 71.7% (95% CI, 58.6 to 82.5) of patients experienced a partial or complete response in cohorts 1, 2, and 3, respectively. In the KEYNOTE-204 study, more partial or completes responses were observed in the pembrolizumab arm relative to the BV arm (65.6%; 95% CI, 57.4 to 73.1 versus 54.2; 95% CI, 46.0 to 62.3), which was associated with a statistically insignificant 11.3% (95% CI, 0.2 to 22.1) difference in favour of pembrolizumab.

Complete Response Rate

In the KEYNOTE-051 study, 28.6% of patients (95% CI, 3.7 to 71.0) experienced a complete response. In the KEYNOTE-087 study, 26.1% (95% CI, 16.3 to 38.1), 25.9 (95% CI, 16.8 to 36.9), and 31.7% (95% CI, 20.3 to 45.0) of patients in cohorts 1, 2, and 3, respectively, experienced a complete response. In the KEYNOTE-204 study, the complete response rate was comparable between the pembrolizumab (24.5%; 95% CI, 17.9 to 32.2) and BV arms (24.2; 95% CI, 17.6 to 31.8).

Duration of Response

In the KEYNOTE-051 study, median DOR was not reached. In the KEYNOTE-087 study, the median DOR in cohorts 1, 2, and 3 were 25.0 months (range = 0 to 36.1), 11.1 months (range = 0 to 35.9), and 16.8 months (range = 0 to 39.1), respectively. In the KEYNOTE-204 study, the median DOR was higher among patients in the pembrolizumab arm (20.7 months; range = 0 to 33.2) than in patients in the BV arm (13.8 months; range = 0 to 33.9).

Time to Response

Median time to response in the KEYNOTE-051 study was 2.6 months (range = 2.1 to 2.8). The median time to response in cohort 1, cohort 2, and cohort 3 of the KEYNOTE-087 study were 2.7 months (range = 2.1 to 12.9), 2.8 months (range = 2.2 to 11.0), and 2.8 months (range = 2.6 to 16.5), respectively. Finally, the median time to response in the pembrolizumab arm of the KEYNOTE-204 study was 2.8 months (range = 1.0 to 31.2) and also 2.8 months (range = 1.3 to 7.3) in the BV arm.

Health-Related Quality of Life

HRQoL data were only measured in the KEYNOTE-087 and KEYNOTE-204 studies. In the KEYNOTE-087 study, the least squares mean change in the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30) global health status between week 24 and baseline was 11.8, 13.9, and 6.6 in cohorts 1, 2, and 3, respectively. No CIs were reported in the KEYNOTE-087 study. In the KEYNOTE-204 study, the least squares mean change in EORTC QLQ-C30 global health status between baseline and week 24 was 8.60 points (95% CI, 3.89 to 13.31) higher in the pembrolizumab arm versus the BV arm. Consistent results were reported for the EORTC QLQ-C30 physical functioning scale (6.24; 95% CI, 1.87 to 10.62), EuroQol 5-Dimensions 3-Levels questionnaire (EQ-5D-3L) utility score (0.09; 95% CI, 0.04 to 0.14), and EQ-5D-3L visual analogue scale (6.12; 95% CI, 1.91 to 10.34).

Table 2: Summary of Key Efficacy Results From Pivotal and Protocol-Selected Studies

Results

KEYNOTE-051

KEYNOTE-087

KEYNOTE-204

Pembrolizumab

N = 7

Cohort 1

N = 69

Cohort 2

N = 81

Cohort 3

N = 60

Pembrolizumab

N = 151

Brentuximab vedotin

N = 153

Progression-free survival (primary analysis)

Number of events, n (%)

3 (42.9)

43 (62.3)

54 (66.7)

36 (60.0)

81 (53.6)

88 (57.5)

Median PFS, months (95% CI)

11.2

(2.6 to NR)

16.4

(11.3 to 27.6)

11.1

(7.3 to 13.5)

19.4

(8.4 to 22.1)

13.2

(10.9 to 19.4)

8.3

(5.7 to 8.8)

PFS at 12 months, % (95% CI)

27.8 (NR)

61.3 (NR)

43.0 (NR)

53.9 (NR)

53.9

(45.0 to 61.9)

35.6

(26.9 to 44.4)

PFS at 24 months, % (95% CI)

NR (NR)

41.6 (NR)

21.9 (NR)

34.0 (NR)

35.4

(26.2 to 44.6)

25.4

(17.1 to 34.5)

Hazard ratio (95% CI)

NA

NA

0.65 (0.48 to 0.88); P = 0.00271a

Overall survival

Number of events, n (%)

NR

11 (15.9)

14 (16.0)

9 (15.0)

||||

||||

Median OS, months (95% CI)

NR (NR)

Not reached

Not reached

Not reached

||||

||||

OS at 12 months, % (95% CI)

100 (NR)

95.7 (NR)

96.2 (NR)

96.6 (NR)

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

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

OS at 24 months, % (95% CI)

NR (NR)

92.6 (NR)

91.0 (NR)

89.4 (NR)

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

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

Hazard ratio (95% CI)

NA

NA

||||

Objective response rate

Number of responses, n (%)

3 (42.9)

54 (78.3)

52 (64.2)

43 (71.7)

99 (65.6)

83 (54.3)

   Complete response

2 (28.6)

18 (26.1)

21 (25.9)

19 (31.7)

37 (24.5)

37 (24.2)

   Partial response

1 (14.3)

36 (52.2)

31 (38.3)

24 (40.0)

62 (41.1)

46 (30.1)

   Stable disease

3 (42.9)

8 (11.6)

8 (9.9)

7 (11.7)

21 (13.9)

36 (23.5)

   Progressive disease

1 (14.3)

5 (7.2)

19 (23.5)

10 (16.7)

26 (17.2)

28 (18.3)

   Not evaluable

0 (0.0)

NR

NR

NR

1 (0.7)

1 (0.7)

   No assessment

0 (0.0)

2 (2.9)

2 (2.5)

0 (0.0)

4 (2.6)

5 (3.3)

Proportion with a response, % (95% CI); P value

42.9

(9.9 to 81.6)

78.3

(66.7 to 87.3)

P < 0.001a

64.2

(52.8 to 74.6)

P < 0.001a

71.7

(58.6 to 82.5)

P < 0.001a

65.6

(57.4 to 73.1)

54.2

(46.0 to 62.3)

Difference in response rate (95% CI); P value

NA

NA

11.3 (0.2 to 22.1); P = 0.022534b

Proportion with complete response, % (95% CI)

28.6

(3.7 to 71.0)

26.1

(16.3 to 38.1)

25.9

(16.8 to 36.9)

31.7

(20.3 to 45.0)

24.5

(17.9 to 32.2)

24.2

(17.6 to 31.8)

Duration of response

Median duration, months (range)

Not reached

(0.0 to 6.1)

25.0

(0 to 36.1)

11.1

(0 to 35.9)

16.8

(0 to 39.1)

20.7

(0.0 to 33.2)

13.8

(0.0 to 33.9)

Patients with extended duration of response, n (%)

   ≥ 6 months

1 (50.0)

34 (81.1)

23 (68.9)

27 (72.7)

66 (79.9)

34 (59.6)

   ≥ 12 months

NR

27 (66.5)

14 (44.4)

19 (64.1)

48 (62.4)

23 (50.0)

   ≥ 18 months

NR

NR

NR

NR

31 (53.7)

13 (42.8)

   ≥ 24 months

NR

15 (50.4)

10 (34.2)

11 (49.8)

11 (47.4)

7 (42.8)

Time to response

Mean, months (SD)

2.5 (0.4)

3.4 (1.8)

3.2 (1.4)

4.2 (2.8)

3.7 (3.9)

2.9 (0.6)

Median, months (range)

2.6

(2.1 to 2.8)

2.7

(2.1 to 12.9)

2.8

(2.2 to 11.0)

2.8

(2.6 to 16.5)

2.8

(1.0 to 31.2)

2.8

(1.3 to 7.3)

Health-related quality of lifec

LS mean change in EORTC QLQ-C30 Global Health Status between week 24 and baseline, mean (95% CI)

NR

11.8 (NR to NR)d

13.9 (NR to NR)e

6.6 (NR to NR)f

7.29 (3.94 to 10.64)

–1.31 (–5.17 to 2.55)

Difference in LS mean change on EORTC QLQ-C30 Global Health Status, (95% CI)

NR

NR

8.60 (3.89 to 13.31)

LS mean change in EORTC QLQ-C30 Physical Functioning Scale between week 24 and baseline, mean (95% CI)

NR

NR

4.31 (1.15 to 7.47)

–1.93 (–5.44 to 1.58)

Difference in LS mean change on EORTC QLQ-C30 Physical Functioning Scale, (95% CI)

NR

NR

6.24 (1.87 to 10.62)

LS mean change in EQ-5D-3L Utility Score between week 24 and baseline, mean (95% CI)

NR

NR

0.04 (0.00 to 0.08)

–0.05 (–0.09 to –0.01)

Difference in LS mean change on EQ-5D-3L Utility Score, (95% CI)

NR

NR

0.09 (0.04 to 0.14)

LS mean change in EQ-5D-3L visual analogue scale between week 24 and baseline, mean (95% CI)

NR

NR

8.53 (5.42 to 11.64)

2.41 (–1.05 to 5.87)

Difference in LS mean change on EQ-5D-3L visual analogue scale, mean (95% CI)

NR

NR

6.12 (1.91 to 10.34)

CI = confidence interval; EORTC QLQ-C30 = European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30; EQ-5D-3L = EuroQol 5-Dimensions 3-Levels questionnaire; LS = least squares; NA = not available; NR = not reported; OS = overall survival; PFS = progression-free survival; SD = standard deviation.

aStatistically significant.

bStatistically insignificant.

cOnly 146 and 150 pembrolizumab and brentuximab vedotin patients, respectively, had complete EORTC data.

d69 individuals had complete data.

eOnly 79 individuals had complete data.

fOnly 58 individuals had complete data.

Source: Clinical Study Reports for the KEYNOTE-051, 87, and 204 studies.

Harms Results

In the KEYNOTE-051 study, 85.7% of patients experienced at least 1 AE. In the KEYNOTE-087 study, 98.6%, 98.8%, and 95.0% of patients experienced at least 1 AE in cohort 1, cohort 2, and cohort 3, respectively. In the KEYNOTE-204 study, 98.0% of patients in the pembrolizumab arm and 94.1% of those in the BV arm experienced an AE. The most common AEs were pyrexia, vomiting, headache, abdominal pain, anemia, cough, fatigue, diarrhea, and upper respiratory tract infections. In the KEYNOTE-204 study, pembrolizumab patients were more likely than BV patients to experience endocrine disorders (20.3% versus 3.9%); infections (66.2% versus 45.4%); musculoskeletal and connective tissue disorders (37.8% versus 31.6%); neoplasms (7.4% versus 1.3%); renal or urinary disorders (14.9% versus 4.6%); respiratory, thoracic, or mediastinal disorders (45.3% versus 26.3%); and skin and subcutaneous tissue disorders (43.9% versus 36.8%), but less likely to experience blood or lymphatic system disorders (18.2% versus 25.7%), gastrointestinal disorders (43.9% versus 52.0%), and nervous system disorders (26.4% versus 50.7%).

In the KEYNOTE-051 study, 28.6% of patients experienced at least 1 serious AE. In the KEYNOTE-087 study, 21.7%, 22.2,% and 25.0% of patients experienced a serious AE in cohort 1, cohort 2, and cohort 3, respectively. In the KEYNOTE-204 study, 29.7% of pembrolizumab and 21.1% of BV-treated patients experienced a serious AE. The most common serious AEs in the KEYNOTE-051 study were diaphragmatic hernia and pneumonia. The most common serious AEs in cohort 1 of KEYNOTE-087 were pneumonia and pericarditis. The most common serious AE in cohort 2 of the KEYNOTE-087 study was herpes zoster and the most common serious AEs in cohort 3 of the KEYNOTE-087 study were pyrexia and pneumonitis, There were no notable differences in frequency of serious AEs between the pembrolizumab and BV arms in the KEYNOTE-204 study. The most common serious AEs in the pembrolizumab arm of the KEYNOTE-204 study were infections or infestations; respiratory, thoracic, or mediastinal disorders; neoplasms; general disorders or administration site conditions; and hepatobiliary disorders. The most common serious AEs in the BV arm of the KEYNOTE-204 study were infections or infestations; respiratory, thoracic, or mediastinal disorders; nervous system disorders; gastrointestinal disorders; and general disorders or administration site conditions.

No patients in the KEYNOTE-051 study discontinued treatment due to an AE, while 11.6%, 6.2%, and 8.3% of patients in cohort 1, cohort 2, and cohort 3, respectively, of the KEYNOTE-087 study discontinued treatment due to an AE. In the KEYNOTE-204 study, 13.5% and 17.8% of patients receiving pembrolizumab and BV discontinued treatment due to an AE, respectively.

In the KEYNOTE-051 study, 28.6% of patients experienced at least 1 immune-mediated AE. In cohort 1, 2, and 3 of the KEYNOTE-087 study, 31.9%, 32.1%, and 38.3% of patients, respectively, experienced at least 1 immune-mediated AE. In the KEYNOTE-204 study, more patients in the pembrolizumab arm (35.8%) than the BV arm (13.8%) experienced an immune-mediated AE. No patients in the KEYNOTE-051 study experienced a serious immune-mediated AE. In the KEYNOTE-087 study, 4.3%, 2.5%, and 5.0% of patients in cohort 1, cohort 2, and cohort 3, respectively, experienced a serious immune-mediated AE. In the KEYNOTE-204 study, more pembrolizumab- than BV-treated patients experienced a serious immune-mediated AE (8.8% versus 3.3%).

Table 3: Summary of Key Harms Results From Pivotal and Protocol-Selected Studies

Harms results, n (%)

KEYNOTE-051

KEYNOTE-087

KEYNOTE-204

Pembrolizumab

N = 7

Cohort 1

N = 69

Cohort 2

N = 81

Cohort 3

N = 60

Pembrolizumab

N = 148

Brentuximab vedotin

N = 152

Patients with at least 1 adverse event

6 (85.7)

68 (98.6)

80 (98.8)

57 (95.0)

145 (98.0)

143 (94.1)

Patients with at least 1 serious adverse event

2 (28.6)

15 (21.7)

18 (22.2)

15 (25.0)

44 (29.7)

32 (21.1)

Patients who discontinued treatment due to adverse event

0 (0.0)

8 (11.6)

5 (6.2)

5 (8.3)

20 (13.5)

27 (17.8)

Patients who died

0 (0.0)

0 (0.0)

2 (2.5)

1 (1.7)

3 (2.0)

2 (1.3)

Patients with at least 1 immune-mediated adverse event

2 (28.6)

22 (31.9)

26 (32.1)

23 (38.3)

53 (35.8)

21 (13.8)

Patients with at least 1 grade III to V immune-mediated adverse event

0 (0.0)

3 (4.3)

4 (4.9)

1 (1.7)

11 (7.4)

5 (3.3)

Patients with at least 1 serious immune-mediated adverse event

0 (0.0)

3 (4.3)

2 (2.5)

3 (5.0)

13 (8.8)

5 (3.3)

Critical Appraisal

The KEYNOTE-051 and KEYNOTE-087 studies were single-arm, open-label trials, while the KEYNOTE-204 study was an open-label, randomized controlled trial. The single-arm trials will be unable to provide definitive evidence of a medication’s superiority over the standard of care while the open-label design of all trials puts them at risk of bias in either direction. However, some bias from the open-label design would be attenuated by the fact that tumour progression was assessed by an independent and blinded assessor in all 3 trials. Further, the randomized nature of the KEYNOTE-204 study will balance prognostic factors at the beginning of the study. The KEYNOTE-204 study permitted patients to be treated with a subsequent anticancer medication following discontinuation of the trial medication (pembrolizumab or BV) which may obscure the trial medication’s true impact on OS. Patients originally randomized to pembrolizumab were permitted to be subsequently treated with BV and vice versa. Almost all patients randomized to BV (97.4%) received a subsequent anticancer therapy while 70.2% of pembrolizumab-treated patients did so. Those randomized to BV were more likely to cross over and subsequently receive pembrolizumab (17.8% subsequently received pembrolizumab versus 1.4% of patients originally randomized to pembrolizumab retreated with BV). Those originally randomized to BV were also more likely to receive nivolumab (19.7%) relative to those randomized to pembrolizumab (3.4%). Finally, 25.0% of patients originally randomized to pembrolizumab received BV while 4.6% of patients originally randomized to BV were retreated with BV. The KEYNOTE-051 study identified 7 pediatric patients with refractory or relapsed cHL which is insufficient to be representative of the true treatment effect in children with this condition. Moreover, it is unclear if these patients failed or were ineligible for salvage chemotherapy and ASCT which is the population of interest in this review. Due to the methodological limitations of the KEYNOTE-051 study, the evidence base is limited to the KEYNOTE-087 and KEYNOTE-204 studies. While the KEYNOTE-204 study is methodologically superior to the KEYNOTE-087 study due to the randomized active control design of the KEYNOTE-204 study, only 1 active control (BV) was tested. The KEYNOTE-087 and KEYNOTE-204 studies excluded individuals with a ECOG status of 2 or greater which could limit its generalizability. Similarly, the KEYNOTE-204 study only compared pembrolizumab to BV. Notably, CADTH reviewed the use of BV in adults with HL after failure of at least 2 multi-agent chemotherapy regimens who are not candidates for ASCT and did not recommend reimbursement.35 However, the clinical experts consulted by CADTH confirmed that in jurisdictions where it is funded, BV is still standard of care due to the lack of superior alternatives. This is in part supported by more recent evidence suggesting the efficacy of BV as third-line therapy in patients who have not received a stem cell transplant.36

Conclusions

The body of evidence included in this review suggests that, when compared to BV, pembrolizumab provides statistically and clinically significant improvement in PFS as well as clinically significant improvements in || ORR, DOR, and HRQoL. |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||||||||||||||| Patients who received BV were generally less likely to experience AEs, serious AEs, or immune-mediated AEs but more likely to discontinue therapy due to an AE. A definitive explanation of this phenomenon cannot be derived from this evidence alone. However, 1 explanation could be that BV-treated patients expected or observed worse health outcomes and thus were less willing to tolerate AEs, even if the rates were lower than in the pembrolizumab arm. Discontinuation would be a viable alternative for these patients as receiving another anticancer medication, including pembrolizumab, was an option. Conversely, pembrolizumab patients may have been willing to tolerate more AEs as the expected benefits were commensurately higher. The body of evidence primarily evaluated pembrolizumab administered 200 mg every 3 weeks in adults but due to the nature of the disease, CADTH’s clinical experts believe that the benefits observed in adults would also be applicable to pediatric patients. However, because of insufficient evidence on the use of pembrolizumab in pediatric patients, it is uncertain what dose should be used to ascertain the benefits observed in adults. No other comparators to pembrolizumab aside from BV were evaluated in the included studies; thus, the comparative effect of pembrolizumab to other relevant treatments in the population under review, beyond BV, remains uncertain. Also, the KEYNOTE-087 and KEYNOTE-204 studies only recruited patients with an ECOG score of 0 or 1 but the CADTH clinical experts did not recommend limiting the use of pembrolizumab only to patients with low ECOG scores. In totality, the evidence suggests that pediatric and adult patients with relapsed or refractory cHL who failed ASCT or are ineligible for multi-agent salvage chemotherapy and ASCT are more likely to benefit from pembrolizumab than from BV; however, the dose required to ascertain these benefits in pediatrics is uncertain.

Introduction

Disease Background

The purpose of this report is to summarize the evidence regarding the use of pembrolizumab monotherapy in adult and pediatric patients with refractory or relapsed cHL who have failed ASCT or who are not candidates for salvage chemotherapy and ASCT. Pembrolizumab is an immune checkpoint inhibitor dosed at 200 mg every 3 weeks in adults and 2 mg/kg every 3 weeks in pediatrics.

The term HL refers to a group of lymphoid proliferations that share clinical and morphological features that distinguish them from other types of lymphoma. It is estimated that in 2020, 1,000 Canadians were diagnosed with cHL and 100 died from the disease.1 Clinically, HL presents most commonly with enlarged cervical lymph nodes, and spread is generally between contiguous nodal areas. Mediastinal masses and B symptoms (fever, weight loss, and night sweats) are common. A bimodal age distribution is appreciated for HL, with most patients diagnosed between the ages of 15 years to 39 years. A second peak is seen in individuals older than 70 years.2 HL is diagnosed by biopsy of an affected tissue or organ. On histopathology large, atypical, and malignant cells, termed Reed-Sternberg or Hodgkin cells, are observed in a heterogeneous background consisting of non-neoplastic inflammatory cells.3 Subclassification of HL into cHL (nodular sclerosis, mixed cellularity, lymphocyte rich, and lymphocyte depleted) and nodular lymphocyte-predominant HL is based on the degree of atypia of the malignant cells, their immunophenotype, and the features of the inflammatory background.

Standards of Therapy

The treatment of cHL is guided by careful assessment of stage- and disease-specific risk factors.19 F-fluorodeoxyglucose PET is considered the gold standard for staging of HL and the Cotswold modification of the Ann Arbor staging system is applied to determine the numerical stage. In 1998 Hasenclever et al. published a prognostic score for adult patients with advanced HL consisting of 7 clinical (sex, age, and stage) and laboratory (anemia, leukocytosis, reduced serum albumin, and lymphopenia) factors. This score predicts freedom from progression (84% for patients with no risk factors to 42% for those with 5 or more factors) and OS (89% for patients with no risk factors to 56% for those with 5 or more factors) at 5 years.4 Treatment is risk-adapted, with low-risk patients (stage I to II, few risk factors) receiving a limited number of cycles of ABVD (doxorubicin-bleomycin-vinblastine-dacarbazine) chemotherapy, often with low-dose involved field radiotherapy5,6 while those with higher-risk disease receiving more extensive chemotherapy. Intensive chemotherapy, in the form of escalated BEACOPP (bleomycin-etoposide-doxorubicin-cyclophosphamide-vincristine-procarbazine-prednisone), is reserved for patients with the highest risk of adverse outcome.7 Response-adapted therapy with the use of interim PET restaging is used to de-escalate treatment for patients who are likely cured8,9 and to escalate treatment of patients who are not responding as expected.8,10 Risk- and response-adapted combined modality therapies are similarly used in pediatric protocols with some differences in chemotherapy backbones.11 Recognizing the shared clinic-pathological features of HL in the most affected age groups, North American adult and pediatric study groups have amalgamated efforts to study new agents using a common chemotherapy backbone.12 The outcome of HL has improved significantly over time and today more than 80% of patients with cHL are cured with initial therapy.2,13

Most patients who do not respond to or who relapse after first-line treatment for HL are treated with high-dose chemotherapy and autologous cell transplantation.14 This approach is supported by 2 randomized trials,15,16 several phase II17-20 and registry21 studies, and results in PFS in 50% to 60% of patients and OS in 60% to 80% of patients. Careful patient selection is required for successful ASCT as the presence of multiple or severe comorbidities may make the treatment-related mortality of high-dose therapy prohibitive. Patients may also not undergo ASCT if they fail to mobilize sufficient numbers of hematopoietic stem cells to support the use of high-dose chemotherapy, if they fail to respond to salvage chemotherapy, or for reasons of conscience as in the case of Jehovah’s Witnesses. Overall, approximately 85% of patients with relapsed or refractory cHL undergo ASCT.

There is currently no standard of care for patients with cHL who relapse after ASCT or who are ineligible for ASCT for 1 of the reasons noted above. Options to treat these patients include chemotherapy, radiotherapy (for those with localized recurrences), targeted therapy with BV, and immune checkpoint inhibitors. Although responses to standard-dose chemotherapy occur frequently in later lines of treatment, the use of conventional-dose salvage chemotherapy is unlikely to lead to cure in these patients.22 Good palliation can be achieved with oral single-agent or combination chemotherapy regimens.23,24 BV is an antibody-drug conjugate that targets CD30-positive cells, delivering the antimitotic agent MMAE into the cytoplasm of these cells by endocytosis.25 The utility of BV in relapsed or refractory cHL was demonstrated in a pivotal phase II study in 102 patients who failed ASCT. The overall response rate was 75% with complete responses seen in 34% of patients and toxicity was manageable, although peripheral neuropathy was frequently dose limiting.26 After 5 years of follow-up, OS in this cohort of 102 patients was 41% and PFS was 22%: Median OS and PFS were higher among those patients who achieved complete remission to BV.27 Real-world experience with BV in transplant-ineligible relapsed or refractory cHL was provided in a phase II study of 136 patients with a median age of 70 years at diagnosis. The most common reasons for transplant ineligibility in this cohort were comorbidities and age. A median of 8 cycles was given and overall and complete responses were observed in 74.3% and 34.6% of patients, respectively, similar to the results seen in patients who had previously undergone ASCT. Median progression-free and OS were 15.1 and 17.8 months, respectively.28

Immune checkpoint inhibitors affect the PD1/PD-L1 axis and lead to increased immune reactivity against cancers that have exploited this mechanism to escape immune control. Nivolumab is a human IgG monoclonal antibody that targets PD1. Nivolumab is licensed for treatment of patients with advanced hepatocellular carcinoma, non–small cell lung cancer, advanced renal cell carcinoma, and certain cases of colorectal carcinoma or malignant melanoma. It is also approved for treatment of patients with cHL who have progressed after ASCT and BV or 3 or more lines of systemic therapy including hematopoietic cell transplantation.29 The indication in cHL is based on the results of the CHECKMATE-205 and CHECKMATE-039 studies, which enrolled a total of 95 patients, demonstrating an overall response rate of 66% and complete and partial remission of 6% and 60%, respectively. Median DOR was 13.1 months. Toxicity was manageable, although immune-mediated toxicity was observed.30,31 A second immune checkpoint inhibitor, pembrolizumab, is the subject of this CADTH review.

Drug

The medication under review is pembrolizumab 200 mg in adults or 2 mg/kg in pediatrics, as monotherapy, administered intravenously every 3 weeks for patients with cHL who have failed ASCT or are ineligible for multi-agent salvage chemotherapy and ASCT. This indication is consistent with the Notice of Compliance provided by Health Canada. This medication has not been assessed by CADTH for this indication in the past.

Table 4: Key Characteristics of Pembrolizumab and Comparators

Characteristics

Pembrolizumab

Brentuximab vedotin

Mechanism of action

Reactivates tumour-specific cytotoxic T lymphocytes in the tumour microenvironment

Disruption of the microtubule network within the cell, leading to cell cycle arrest and cell death

Indicationa

Treatment of adult and pediatric patients with refractory or relapsed cHL, as monotherapy, who have failed ASCT, or who are not candidates for multi-agent salvage chemotherapy and ASCT

Treatment of patients with cHL after failure of ASCT or after failure of at least 2 multi-agent chemotherapy regimens in patients who are not ASCT candidates

Route of administration

IV

IV

Recommended dose

200 mg (adults) or 2 mg/kg (pediatrics) every 3 weeks

1.8 mg/kg every 3 weeks

Serious adverse events

Immune-mediated adverse events

Neurotoxicity

ASCT = autologous stem cell transplant; cHL = classical Hodgkin lymphoma.

aHealth Canada–approved indication.

Source: Health Canada drug product monographs for pembrolizumab37 and brentuximab vedotin.38

Stakeholder Perspectives

Patient Group Input

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

One patient group, Lymphoma Canada, provided input for the review of pembrolizumab for adult and pediatric patients with refractory or relapsed cHL. The indication under consideration is pembrolizumab as monotherapy, for those who have failed ASCT, or who are not candidates for salvage chemotherapy and ASCT. Lymphoma Canada is a national charity that collaborates with patients, caregivers, health care professionals, and other stakeholders to empower the lymphoma community.

Lymphoma Canada conducted 2 anonymous, online surveys of patients with HL. One survey was conducted from June 5 to 30, 2017, to which 91 patients responded. No caregivers responded. The other survey was conducted from November 6, 2020, to January 13, 2021, to which 37 patients responded. The survey link was provided via email to patients registered with Lymphoma Canada, and made available via social media platforms, HL-specific forums, and social media groups, Canadian and American Cancer Society message boards, and with physicians at Canadian clinical trial sites. The survey included multiple-choice questions, rating questions, and open-ended questions. Three patients with HL in Canada, who had direct experience with pembrolizumab, were interviewed over the phone.

Not all respondents provided demographic information (103 out of 128 provided this information for country, and 94 out of 128 provided age and gender information). Of those that did provide demographic information, the majority (55%) were from Canada. Of the 9 patients with pembrolizumab experience, 7 reside in Canada, and 2 reside in the US. Table 5 details the country of survey respondents. Of those that provided demographic information, most respondents are male (54%). Most patients are between the ages of 20 to 59 years of age (78%). Table 6 details the gender and age of the survey respondents.

Table 5: Country of Survey Respondents (128 Respondents)

Respondents

Canada

US

UK

European Union

Other

Skipped

Total

Patients WITHOUT pembrolizumab experience

64

4

12

6

8

25

119

Patients WITH pembrolizumab experience

7

2

0

0

0

0

9

Table 6: Gender and Age of Survey Respondents

Respondents

Age range (years)

Gender

< 20

20 to 39

40 to 59

≥ 60

Skipped

Female

Male

Skipped

Patients WITHOUT pembrolizumab experience

3

38

29

15

34

60

25

34

Patients WITH pembrolizumab experience

0

7

0

2

0

3

6

0

Disease Experience

Experience at Diagnosis

Most of the survey respondents were between the ages of 13 years to 39 years when they were diagnosed (63%; 80 of 128 respondents). When asked about their experience receiving their diagnosis, 11% of respondents did not have all their questions answered, and 27% did not know what questions to ask their doctor who did not explain the disease to them (81 respondents). Some patients commented:

At diagnosis, the HL symptoms that most affected respondents’ quality of life were (based on the responses of 97 patients):

Other symptoms affecting the quality of life for greater than 10% of respondents included loss of appetite, trouble breathing, fever and chills, and chest pain.

When asked about the negative mental and emotional impacts of their disease and treatment, which affected their quality of life at diagnosis, most patients had 1 or more negative impacts (based on the responses of 97 patients):

Current Experience

Thirty-five patients responded to questions about their current symptoms and quality of life. Current symptom experience, for those that responded, included the following:

In all, 113 patients provided information on current social and psychological impacts. These include the following:

Table 7 describes the negative impact of HL on the quality of life of patients.

Table 7: Effect of HL on Day-to-Day Life of Patients (109 Respondents)

Aspect of life negatively impacted by HL

Respondents, n (%)

Ability to work

70 (64)

Personal image

64 (59)

Family

59 (54)

Friendships

49 (45)

Intimate relations

52 (48)

Ability to attend school

19 (17)

HL = Hodgkin lymphoma.

Three patients describe their experience with HL this way:

Experiences With Currently Available Treatments

Of the 85 patients who provided information on previous treatments, all had received treatment or are currently undergoing treatment. Most (94%) had received at least 1 line of conventional chemotherapy, and 24% had received 3 or more lines of therapy. Regarding chemotherapy, the most common regimen received was ABVD (85%). GDP (gemcitabine, dexamethasone, cisplatin) was the next most common (11%), followed by BEACOPP (7%), and least common was BV (6%).

Regarding other treatments, 83 patients provided information, of which 49% received radiation therapy, 20% had an ASCT, and 25% had surgery.

In all, 101 patients indicated their current treatment phase. Following their most recent line of therapy, 85% of respondents are in remission, of which 32% have been in remission for longer than 5 years.

Many respondents are concerned with toxicity and side effects of previous treatments. Table 8 details the most common side effects experienced by respondents during their treatments for HL.

Table 8: Side Effects of HL Treatments (90 Respondents)

Side effect

Respondents, n (%)

Fatigue

86 (96)

Hair loss

83 (92)

Nausea/vomiting

80 (89)

Mouth sores

56 (62)

Anemia and/or neutropenia

59 (66)

Peripheral neuropathy

52 (58)

Low platelets

39 (43)

Diarrhea

38 (42)

Cough or breathing difficulties

44 (49)

Back/joint pain

34 (38)

Skin rashes/severe itching

32 (36)

Loss of menstrual periods

27 (30)

Infections

25 (28)

HL = Hodgkin lymphoma.

When asked which side effects patients found most difficult to tolerate, respondents reported nausea/vomiting (43%), fatigue (41%), hair loss (13%), mouth sores (10%), and bowel obstruction (4%) (68 respondents). Long-lasting side effects of treatments, reported lasting longer than 2 years or appearing 2 years or later after treatment, included fatigue (66%), “chemo brain” (60%), peripheral neuropathy (41%), loss of menstrual periods (18%) and sterility (18%), chest pain or infection (15%), and thyroid problems (20%) (80 respondents).

Table 9 details the impact of treatment on respondents’ quality of life. The question was a scale from 1 to 5, where 1 equals “no impact” and 5 equals “significant negative impact.”

Table 9: Impact of Treatment on Quality of Life (90 Respondents)

Treatment aspect

Weighted average

Number of responses

Fatigue

4.2

90

Infusion reaction

3.3

88

Number of clinic visits

3.2

89

Infusion time

3.0

90

Incidence of infections

2.1

90

Table 10 details the impact of treatment on respondents’ daily living. The same scale as the question detailed in Table 9 was used.

Table 10: Impact of Treatment on Daily Living (90 Respondents)

Activity

Weighted average

Number of responses

Work

3.6

90

Travel

3.5

89

Intimate relations

3.5

90

Family

3.3

90

Friendships

3.3

90

School

1.2

87

When discussing treatments and side effects, 3 respondents reported their experience in this way:

Access to Treatment

Many patients (83% of the 90 individuals who responded) were able to access treatment in their own communities. For the 15 patients who were unable to access treatment in their own communities, 73% lived in a community without a cancer care centre and for 26%, the treatment was not available at the local cancer care centre.

Table 11 details the financial impact of treatment.

Table 11: Financial Implications of Treatment for Patients With HL (86 Respondents)

Financial impact

Respondents, n (%)

Absence from work or school

62 (72)

Parking

39 (45)

Cost of medications

27 (31)

Travel

19 (22)

None

12 (14)

Accommodation

6 (7)

Drug disposal issues

3 (3)

HL = Hodgkin lymphoma.

Improved Outcomes

Patients seek individualized treatment options that provide disease control and remission, with fewer side effects than current treatment options available. When asked about important factors of new drugs or treatments for HL, “longer survival” and “longer remission” were the most important outcomes. Table 12 details the responses from the 24 patients who answered this question. The scale for the question ranged from 1 (“not important”) to 5 (“extremely important”).

Table 12: Patient Treatment Preferences (24 Respondents)

Treatment outcome or factor

% of participants rating question as extremely important (rating = 5)

Weighted average

Longer survival

96

4.96

Longer remission

96

4.88

Better quality of life

92

4.92

Fewer side effects

67

4.46

Of the 89 respondents who were asked if they would be willing to tolerate short-term side effects of a new treatment, 55% would be willing to tolerate potential short-term side effects, while 31% were not; the remaining 14% were unsure. Respondents were also asked if they would choose a treatment with known and potentially serious side effects if their doctor recommended it was the best option for them. Of the 100 patients who answered this question, 53% selected “Yes,” while only 3% selected “No”; the remaining 44% were unsure.

The survey asked respondents how important it is for patients and physicians to have a choice of therapy (a scale of 1 to 10, with 1 being “not important,” and 10 being “very important”). Most participants (79%) rated this a 7, 8, 9, or 10 (weighted average was 8.2).

A scale of 1 (“least important”) to 10 (“most important”) was used to ask survey participants which HL symptoms would be most important for a new treatment to control. One hundred respondents answered this question. Patients rated the most important symptoms for new treatments to control include difficulty breathing (8.1), drenching night sweats (7.2), chest pain (7.6), fatigue/lack of energy (7.4), and enlarged spleen or abdominal discomfort (7.0).

Experience With Drug Under Review

Nine patients had experience with pembrolizumab; 3 of these patients were interviewed for the patient input submission. The reasons for starting treatment with pembrolizumab include: no other treatment options were available (2 patients); HL progressed after autologous transplant and did not want to risk the potential toxicity of an allogeneic transplant (4 patients), hoping for remission to proceed to allogenic transplant (1 patient); did not respond to 3 previous lines of chemotherapy, and did not want to undergo an autologous transplant (2 patients).

Table 13 details the patients with pembrolizumab experience.

Table 13: Patients With HL With Pembrolizumab Experience

Patient

Gender

Age (years)

Location

Year of diagnosis

Access to drug

Year started pembrolizumab

1

Male

20 to 39

US

2011

Clinical trial

Not reported

2

Male

31

Canada

2014

Clinical trial

2016

3

Male

24

Canada

2016

Clinical trial

2017

4

Female

20 to 39

US

2014

Private insurance

Not reported

5

Female

27

Canada

2010

Clinical trial

2015

6

Female

60 to 69

Canada

NA

NA

2018

7

Male

31 to 39

Canada

2012

Clinical trial

2017

8

Male

70 to 79

Canada

2014

Clinical trial

2017

9

Male

20 to 25

Canada

2017

NA

2020

HL = Hodgkin lymphoma; NA = not available.

All 9 patients had at least 2 prior lines of conventional chemotherapy, and 3 of these patients had received 6 or more lines of therapy. Previous chemotherapy treatments included ABVD (8), GDP (6), GVD (2), COPP (1), DHAP (1), bendamustine (1), lenalidomide (1), and unknown (1). Regarding other therapies, 7 patients had undergone an ASCT, 1 had undergone an allogeneic stem cell transplant and 4 had received treatment with BV before beginning treatment with pembrolizumab. When asked which symptoms pembrolizumab managed, 7 of the 9 patients responded that pembrolizumab managed all their HL symptoms, including fatigue, enlarged lymph nodes, frequent infections, weight loss, night sweats, shortness of breath, and pain. Two patients reported that pembrolizumab did not manage their fatigue.

Regarding the side effects of pembrolizumab, 8 patients tolerated this therapy well. However, 1 patient had to stop treatment with pembrolizumab because of toxicity and side effects, including peripheral neuropathy and inflammatory arthritis for which medication was taken. Table 14 outlines the side effects of pembrolizumab.

Table 14: Side Effects Experienced With Pembrolizumab (9 Respondents)

Side effect

Responses, n (%)

Joint pain

4 (44)

Shortness of breath

4 (44)

Diarrhea

3 (33)

Fatigue

3 (33)

Itching/rash

3 (33)

Cough

2 (22)

Nausea

2 (22)

Other (fever)

2 (22)

None of these

1 (11)

Peripheral neuropathy

1 (11)

Knowing the potential side effects, all 9 patients responded that they would take this drug again if their doctor thought it was the best choice. The patient who had to stop treatment due to toxicity stated, “PFS was worth the side effects.”

When asked how the side effects of pembrolizumab compared to other treatments, 3 patients provided comments:

Regarding quality of life and impact on daily activities, 7 of the 9 patients reported that they did not experience any negative impact on work or school, family obligations, friendships, intimate relations, activities, or travel. One patient reported lasting fatigue that was thought to have been due to the drug, and the fatigue limited aspects of their life. The patient with lasting side effects of peripheral neuropathy and inflammatory arthritis had this negatively impact their family life and personal image.

Overall Experience With Pembrolizumab

All 9 patients said they had a good to excellent experience with pembrolizumab, and all would take this treatment if offered to them again. Based on their own experience, all 9 patients would recommend this therapy to other patients with HL.

When reflecting on their overall health and well-being, 3 patients had this to say:

Companion Diagnostic Test

There is no companion diagnostic testing for pembrolizumab.

Clinician Input

Input From Clinical Experts Consulted by CADTH

All CADTH review teams include at least 1 clinical specialist with expertise regarding the diagnosis and management of the condition for which the drug is indicated. Clinical experts are a critical part of the review team and are involved in all phases of the review process (e.g., providing guidance on the development of the review protocol, assisting in the critical appraisal of clinical evidence, interpreting the clinical relevance of the results, and providing guidance on the potential place in therapy). The following input was provided by 3 clinical specialists with expertise in the diagnosis and management of pediatric and adult cHL.

Unmet Needs

For pediatric patients, the clinical expert highlighted that the current treatment options are aggressive cytotoxic chemotherapy, which can have serious side effects such as infections and organ toxicities. If patients do not respond to these treatments, there are limited remaining treatment options. The goal for pediatrics is cure of disease whenever possible, as well as minimizing AEs and treatment-related morbidities, improving HRQoL, and delaying disease progression.

The experts who treat adult patients stated several unmet needs in this patient population, mainly that none of the currently available treatments are curative. Current treatments are also limited by their means of administration (e.g., IV administration, which requires a hospital visit) and toxicity associated with treatment. There is a need for better tolerated treatments, formulations to improve convenience, and treatments for patients who do not respond or become refractory to current treatments.

Place in Therapy

The pediatric clinical expert highlighted that studies evaluating the addition of pembrolizumab to upfront and first salvage therapies is highly anticipated. The successful application of pembrolizumab in the relapsed and refractory cHL setting could offer a potential cure, prolonged disease control, and improved quality of life for these patients that are highly pre-treated with other drug therapies.

The adult clinical expert stated that pembrolizumab has the potential to be used earlier in the current treatment paradigm. They highlighted findings that support the use of pembrolizumab after failed ASCT, or as a second-line therapy for patients who are not eligible for salvage chemotherapy or ASCT. Pembrolizumab, while used as a single therapy currently, could potentially be combined with other therapies such as chemotherapy for patients who relapse.

Patient Population

For pediatric patients, the clinical expert suggests that patients meeting criteria, and without comorbidities that would make them ineligible, should be considered for treatment with pembrolizumab. For the adult population, patients with relapsed cHL would be identified by their hematologist or oncologist (their disease state may be apparent by patient symptoms, clinical examination, or medical imaging). Relapsed disease would be confirmed by a biopsy.

It was noted that patients with relapsed and refractory cHL are closely monitored for disease progression. If patients relapse, they are generally offered treatment regardless of whether or not their lymphoma is causing them symptoms; otherwise, their disease will continue to progress. None of the experts were aware of biomarkers that can determine which patients will respond to treatment; in early studies for checkpoint inhibitors, evidence of PD1 or PDL1 expression on biopsy samples was required, but this incidence is almost 100% in patients with cHL and no longer a requirement.

Patients least suited to pembrolizumab, as noted by the pediatric clinical expert, are those patients on therapy or recently on therapy for autoimmune disease, patients with poor performance status, or patients with organ failure. The adult experts highlighted that some patients with localized disease may be better suited for radiation therapy and that patients at high risk for autoimmune side effects with pembrolizumab would be least suited to this treatment.

Assessing Response to Treatment

The pediatric expert commented that a clinically meaningful response to treatment would be resolution of disease symptoms and radiologic evidence of disease response. This response would lead to an allogenic stem cell transplant, if eligible, or to ASCT if previously ineligible, along with improved HRQoL outcomes, and improved ability to perform activities of daily living. Treatment response should be assessed after the first 2 to 4 cycles of therapy (corresponding to 6 to 12 weeks), and then every 12 weeks thereafter.

For adult patients, the clinical experts highlighted improvement in symptoms, radiological evidence of disease response, and reduction in the size of lymph nodes and other disease sites. Goals of treatment would be response (either partial or complete); improved survival; improved quality of life, including ability to perform the activities of daily living and a return to work; and improved symptoms, but ideally complete resolution of symptoms. Patients receiving pembrolizumab are assessed clinically at every visit (every 3 weeks) and radiologically every 3 to 4 cycles.

Discontinuing Treatment

Treatment should be discontinued if there is disease progression, severe immune-related AEs, or severe infusion or hypersensitivity reactions. The pediatric expert noted that checkpoint inhibitors, including pembrolizumab, may be associated with a phenomenon called pseudoprogression, an inflammatory response which does not represent disease progression. Disease response should be carefully considered.

Prescribing Conditions

Pediatric oncologists or adult hematologists or oncologists are required to diagnose, treat, and monitor patients with relapsed or refractory cHL who are receiving pembrolizumab. If a patient experiences an immune-related AE, they may be referred to another specialist. Pembrolizumab can be given in the outpatient setting.

Clinician Group Input

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

Three registered clinician groups provided input for this review. One submission was by the Ontario Health Hematology Disease Site Drug Advisory Committee (Cancer Care Ontario; OH-CCO DAC), which included 6 physicians. OH-CCO DAC provides evidence-based clinical and health system guidance for the Provincial Drug Reimbursement Programs and Systemic Treatment Program. The Lymphoma Canada Scientific Advisory Board provided a separate submission; the group consisted of 5 clinicians. Lymphoma Canada is a not-for-profit organization for Canadian patients with lymphoma and chronic lymphocytic leukemia and more information on this organization can be found at www.lymphoma.ca. POGO also completed a submission; this submission was coordinated by 1 physician, with the input from POGO’s Therapeutic and Technology Advisory Committee. Ontario’s 5 specialized childhood cancer centres, and an official advisor to the Ontario Ministry of Health and Long-Term Care, comprise POGO. More information about POGO can be found at www.pogo.ca.

Unmet Needs

Both Lymphoma Canada and OH-CCO DAC identified highest unmet need in patients who have failed ASCT or are ineligible for ASCT. Lymphoma Canada noted that across the country there are “access gaps” for novel therapies (e.g., BV and anti-PD1 antibodies), due to lack of funding. OH-CCO DAC also echoed that BV is not covered for patients who are transplant ineligible. For patients that are not able to receive ASCT because of a lack of disease response, these patients need an effective therapy to position them to receive ASCT. Lymphoma Canada also stated that standard therapies (e.g., salvage chemotherapy) typically are more toxic and less effective than novel therapies; they also are not associated with favourable PFS or OS, or meaningful long-term disease control. They noted that novel therapies with a favourable efficacy to toxicity ratio are needed.

In the submission from POGO, they identified the greatest unmet need to be patients who have relapsed or refractory cHL and who have had previous exposure to BV. Even in the relapsed or refractory cHL child and adolescent population, the goal of therapy is cure and disease response.

Place in Therapy

In the submission from Lymphoma Canada, pembrolizumab, as supported by the KEYNOTE-204 clinical trial, would be used for patients with relapsed or refractory cHL after primary therapy if they are ineligible for ASCT, and in other patients who have received at least 2 prior lines of therapy, or have relapsed after ASCT. This is in-line with using pembrolizumab in second-line or beyond. It is anticipated that clinical practice would change based on the data from the KEYNOTE-204 study; for example, pembrolizumab would be used instead of BV in the post-ASCT population, or for patients who are not eligible for ASCT and have received prior therapy. It is thought that pembrolizumab might replace BV regarding place in therapy, for patients with relapsed or refractory cHL (funding for BV is limited).

The submission from OH-CCO DAC identified that younger patients who failed first-line therapy and do not respond to salvage chemotherapy (i.e., are ineligible for ASCT), would likely receive pembrolizumab. Older patients who fail first-line therapy and are not eligible for ASCT due to comorbidities or age would likely receive pembrolizumab instead of salvage chemotherapy.

For the pediatric population, where many patients have had past exposure to BV, pembrolizumab would be used after BV. Pembrolizumab would be appropriate for patients who have already received an ASCT and BV, or who fail to respond to BV or experience toxicity.

Patient Population

Both OH-CCO DAC and Lymphoma Canada identified patients who met the criteria outlined by the KEYNOTE-204 clinical trial as experiencing the greatest unmet need for patients with relapsed or refractory cHL, and most suited to receive pembrolizumab. Lymphoma Canada specifically noted adult patients who experienced failure of primary treatment, or who have not responded to second-line treatment. They also identified children and adolescents with relapsed or refractory cHL who have experienced failure after ASCT. Lymphoma Canada stated that most patients with relapsed or refractory cHL would be eligible for pembrolizumab. POGO identified patients who have progressive or relapsed disease after BV, or who are unable to tolerate it, but have acceptable performance status (ECOG 0 or 1 or Lansky Performance Scale score > 60) as most in need of and best likely to tolerate pembrolizumab.

Assessing Response to Treatment

To assess treatment response, Lymphoma Canada identified that patients typically undergo serial imaging (i.e., fluorodeoxyglucose PET) to monitor disease progression, regardless of ASCT eligibility. POGO also identified cross-sectional imaging and PET scans as the means for determining disease progression and treatment response. OH-CCO DAC stated that disease response is determined by standard response criteria, including imaging. Frequency of imaging varies across the country, but typically for the ASCT-eligible population it has typically been done at 3 months, and 1 year after ASCT. For the ASCT-ineligible population, imaging could be based on patient symptoms or after treatment. A clinically meaningful response to treatment would include improvement in disease related symptoms, tumour response, and disease control (i.e., PFS or OS). POGO and OH-CCO DAC suggested disease status should be assessed every 12 weeks, at minimum.

Discontinuing Treatment

When deciding to discontinue treatment, Lymphoma Canada and POGO indicated that disease progression and significant toxicities (i.e., AEs, particularly grade 3 or 4 events, and immune-related events) would be important considerations.

Prescribing Conditions

The submission from Lymphoma Canada stated that there is evidence to support the use of anti-PD1 antibodies in a variety of malignancies, with administration of treatment in the community setting, hospitals, and tertiary cancer centres. OH-CCO DAC stated that outpatient clinics would be suitable to administer pembrolizumab. For the pediatric population, POGO suggested that pembrolizumab be administered in specialized pediatric cancer programs only.

Drug Program Input

The drug programs provide input on each drug being reviewed through CADTH’s reimbursement review processes by identifying issues that may impact their ability to implement a recommendation. The implementation questions and corresponding responses from the clinical experts consulted by CADTH are summarized in Table 15.

Table 15: Summary of Drug Plan Input and Clinical Expert Response

Drug program implementation questions

Clinical expert response

If a patient who is otherwise eligible refuses to undergo a transplant, would it be appropriate to treat with pembrolizumab?

Yes, based on their expertise, the clinical experts believe such patients would be eligible for pembrolizumab treatment.

If a patient can proceed to transplant after responding to pembrolizumab (in the absence of progressive disease), can the patient be retreated with pembrolizumab upon relapse? If yes, what is the minimum time interval between the transplant and the relapse to be considered for pembrolizumab re-treatment? Are there circumstances wherein re-treatment with pembrolizumab should not be considered?

There is insufficient evidence at this time to support re-treatment in this setting; however, there are some case reports of re-treatment and a recent retrospective case series supportive of the practice in cHL in other settings.39,40

Re-treatment was not noted in the KEYNOTE-204 protocol. If a patient received up to 35 cycles of pembrolizumab every 3 weeks, would the patient be eligible for additional pembrolizumab re-treatment (e.g., 17 cycles)?

In both KEYNOTE-trials, patients were treated with pembrolizumab for a maximum of 24 months (i.e., 35 cycles) or until disease progression, intolerable toxicity, or investigator’s decision. Re-treatment with pembrolizumab was not allowed in KEYNOTE-204. However, KEYNOTE-087 allowed patients to receive an additional 17 cycles of pembrolizumab beyond initial progression if they met the following criteria:

  • Stopped initial trial therapy after having confirmed CR by study investigator

  • Received at least 24 weeks of pembrolizumab before discontinuing treatment and had at least 2 treatments beyond the date of initial CR

  • Had disease progression as assessed by the study investigator after stopping initial pembrolizumab treatment

  • Did not receive any anticancer therapies after initial pembrolizumab therapy

  • Had an ECOG performance status of 0 or 1 and adequate organ function

Patients who had CR and were treated for up to 2 years with pembrolizumab could also receive an additional 17 cycles of pembrolizumab in KEYNOTE-087.

The clinical experts consulted by CADTH agreed that these patients may be eligible for re-treatment with an additional 17 cycles of pembrolizumab upon experiencing disease progression.

Would patients whose disease has progressed on a prior PD-1 or PD-L1 inhibitor be eligible for pembrolizumab.

While there is a case report of pembrolizumab as bridge therapy to ASCT followed by nivolumab at time of relapse after ASCT, the experts were skeptical of this approach as the mechanism of action of pembrolizumab and nivolumab are too similar. Currently, the data are too limited to support this approach.

If there is progression during a drug holiday, can treatment be resumed?

There is no evidence to support resuming treatment after a progression event that occurred during a drug holiday.

Should the following patients who were excluded in the pembrolizumab trials be eligible: adult patients with an ECOG score ≥ 2, pediatric patients with a Lansky Play-Performance Scale score < 50 (for children aged ≤ 16 years) or a Karnofsky score < 50 (for children aged > 16 years).

The clinical experts believed that patients with less favourable performance status can also experience treatment benefit.

For patients currently treated with brentuximab vedotin, are there circumstances where it would be better to continue the brentuximab vedotin and reserve pembrolizumab downstream?

It would depend on the goal of brentuximab therapy, but based on their clinical expertise, CADTH’s clinical experts noted that these patients should continue receiving brentuximab vedotin if they are responding well to the treatment and have not experienced disease progression.

What downstream sequencing options are available after pembrolizumab?

In pediatrics, an allogenic transplant, or enrolment in a phase I clinical trial would be reasonable. In adults, an allogenic or autologous transplant or palliative chemotherapy would be reasonable options.

ASCT = autologous stem cell transplant; cHL = classical Hodgkin lymphoma; CR = complete response; ECOG = Eastern Cooperative Oncology Group.

Clinical Evidence

The clinical evidence included in the review of pembrolizumab is presented as a systematic review which includes pivotal studies provided in the sponsor’s submission to CADTH and Health Canada, as well as those studies that were selected according to an a priori protocol.

Systematic Review (Pivotal and Protocol-Selected Studies)

Objectives

To perform a systematic review of the efficacy and safety of pembrolizumab, as monotherapy, in adult and pediatric patients with refractory or relapsed cHL, who have failed ASCT or who are not candidates for salvage chemotherapy and ASCT.

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 16.

Outcomes included in the CADTH review protocol reflect outcomes considered to be important to patients, clinicians, and drug plans.

Table 16: Inclusion Criteria for the Systematic Review

Criteria

Description

Patient population

Adult and pediatric patients with refractory or relapsed classical Hodgkin lymphoma, as monotherapy, who have failed autologous stem cell transplant or who are not candidates for salvage chemotherapy and autologous stem cell transplant

Intervention

Pembrolizumab 200 mg administered intravenously every 3 weeks in adult patients

Pembrolizumab 2 mg/kg administered intravenously every 3 weeks in pediatric patients

Comparators

Adults:

  • Brentuximab vedotin

  • Nivolumab

  • Radiation therapy

  • Allogenic transplant

  • Bendamustine

  • Lenalidomide

Pediatrics:

  • Brentuximab and gemcitabine

  • Brentuximab and bendamustine

  • GV (gemcitabine and vinorelbine) for those have received brentuximab

  • IGEV (ifosfamide, gemcitabine, and vinorelbine)

  • GDP (gemcitabine, dexamethasone, and cisplatin)

  • ICE (ifosfamide, carboplatin, and etoposide)

  • DHAP (dexamethasone, cytarabine, and cisplatin)

Outcomes

Efficacy outcomes:

  • Overall survival

  • Progression-free survival

  • Event-free survival (for pediatric patients)

  • Overall response rate

  • Complete response rate

  • Duration of response

  • Time to response

  • HRQoL

Harms outcomes:

  • AEs, SAEs, WDAEs, mortality, and immune-mediated AEs

Study design

  • Published and unpublished phase III and IV RCTs

AE = adverse event; HRQoL = health-related quality of life; RCT = randomized controlled trial; SAE = serious adverse event; WDAE = withdrawal due to adverse event.

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.41 Published literature was identified by searching the following bibliographic databases: MEDLINE All (1946‒) via Ovid and Embase (1974‒) via Ovid. The search strategy comprised both controlled vocabulary, such as the National Library of Medicine’s MeSH (Medical Subject Headings), and keywords. The main search concepts were Keytruda (pembrolizumab) and classical HL. Clinical trials registries were searched: the US National Institutes of Health’s clinicaltrials.gov, WHO’s International Clinical Trials Registry Platform 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. See Appendix 1 for the detailed search strategies. The initial search was completed on March 17, 2021. Regular alerts updated the search until the meeting of the CADTH pan-Canadian Oncology Drug Review Expert Committee meeting on July 15, 2021.

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.42 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. See Appendix 1 for more information on the grey literature search strategy. 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 HL was run in MEDLINE All (1946–) on March 17, 2021. No limits were applied to the search.

Findings From the Literature

A total of 359 studies were identified from the literature for inclusion in the systematic review (Figure 1). The included studies are summarized in Table 17. Six reports based on 3 unique studies were identified and were all provided by the sponsor; no additional unique studies were identified from the literature search. No network meta-analyses were identified.

Figure 1: Flow Diagram for Inclusion and Exclusion of Studies

312 citations were identified, 304 were excluded, and 6 additional relevant reports were identified from other sources. Of these, 14 potentially relevant full-text reports were retrieved for scrutiny. In total, 6 reports were included in the review.

Table 17: Details of Included Studies

Detail

KEYNOTE-051

KEYNOTE-087

KEYNOTE-204

Designs and populations

Study design

  • Two-part, nonrandomized, phase I/II, open-label, single-arm, multi-centre study recruiting patients with several types of cancer

  • Part 1 evaluated the dose, safety, pharmacokinetics, pharmacodynamics, toxicity, and preliminary efficacy of pembrolizumab. Part 2 further evaluated safety and efficacy.

Single-arm, nonrandomized clinical trial with 3 cohorts:

  • Patients who failed to respond to or progressed after ASCT and relapsed after or failed to respond to treatment with BV after ASCT

  • Patients who were ineligible for ASCT and relapsed after or failed to respond to BV

  • Patients who failed to respond or progressed after ASCT and had not yet received BV.

Phase III, open-label, randomized controlled trial

Locations

51 centres in 12 countries

51 centres (number of countries not provided)

123 centres in 20 countries

Patient enrolment dates

First patient enrolled on March 23, 2015, with last visit of last patient on January 10, 2020, and a database cut-off of January 10, 2020

First patient visit on June 16, 2015, with data cut-off of March 21, 2019

First patient enrolled on June 29, 2016, with last visit of last patient on January 16, 2020, and a database cut-off of February 17, 2020

Randomized (N)

None

None

304

Inclusion criteria

  • 3 to 18 years of age

  • Have relapsed or refractory cHL and are either: refractory to front-line therapy; or high-risk and relapsed from front-line therapy; or relapsed or refractory to second-line therapy (relapsed was defined as disease progression after most recent therapy and refractory was defined as failure to achieve CR or PR)

  • Have a performance status as: Lansky Play-Performance Scale score ≥ 50 for children 16 years of age and younger; Karnofsky score ≥ 50 for children older than 16 years of age.

  • ≥ 18 years of age

  • Have relapsed or refractory cHL and meet 1 of the following criteria: have failed to achieve a response or progressed after ASCT (patients must have relapsed after treatment with or failed to respond to brentuximab after ASCT); are ineligible (unable to achieve a CR or PR to salvage chemotherapy) for ASCT (patients must have relapsed after treatment with or failed to respond to brentuximab after ASCT); or have failed to achieve a response or progressed after ASCT and have not received BV after ASCT

  • ≥18 years of age

  • Have relapsed (disease progression after most recent therapy) or refractory (failure to achieve CR or PR to most recent therapy) cHL and meet 1 of the following criteria: have failed to achieve a response or progressed after ASCT; or are not ASCT candidates due to chemo-resistant disease (unable to achieve CR or PR on salvage chemotherapy), advanced age (≥ 65 years of age), or any significant coexisting medical condition likely to have a negative impact on tolerability of ASCT

Inclusion criteria

  • Have a performance status of 0 or 1 on the ECOG Performance Scale

  • Patients must have received at least 1 prior multi-agent chemotherapy regimen. Prior treatment with BV or a BV-containing regimen was allowed, provided the participants had responded (PR or CR) to the BV or BV-containing regimen

  • Have a performance status of 0 or 1 on the ECOG Performance Scale

Exclusion criteria

  • Has received prior systemic anticancer therapy including investigational agents within 2 weeks before study day 1 or has not recovered from AEs due to a previously administered agent

  • Has received prior radiotherapy within 2 weeks of start of study treatment; participants must have recovered from all radiation-related toxicities, not require corticosteroids, and not have had radiation pneumonitis

  • Has undergone solid organ transplant at any time, or prior allogeneic hematopoietic stem cell transplantation within the last 5 years

  • Has had a prior monoclonal antibody within 4 weeks prior to study day 1 or has not recovered from AEs due to agents administered more than 4 weeks earlier

  • Has had prior chemotherapy, targeted small molecule therapy, or radiation therapy within 2 weeks prior to study day 1 or has not recovered from AEs due to a previously administered agent

  • Has undergone prior allogeneic hematopoietic stem cell transplantation within the last 5 years

  • Has received prior therapy with an anti-PD-1, anti-PD-L1, anti-PD-L2, anti-CD137, or anti-cytotoxic T-lymphocyte-associated antigen-4

  • Has had a prior monoclonal antibody within 4 weeks prior to first dose of therapy in the study or has not recovered from AEs due to agents administered more than 4 weeks earlier

  • Has had prior chemotherapy, targeted small molecule therapy, or radiation therapy within 2 weeks prior to study day 1 or has not recovered from AEs due to a previously administered agent

  • Has undergone prior allogeneic hematopoietic stem cell transplantation within the last 5 years

  • Has received prior therapy with an anti-PD-1, anti-PD-L1, anti-PD-L2, anti-CD30, anti-CD137, or CTLA-4 antibody (including ipilimumab) or any other antibody or drug specifically targeting T-cell co-stimulation or checkpoint pathways

Drugs

Intervention

Pembrolizumab 2 mg/kg every 3 weeks for 35 cycles or until disease progression, unacceptable AEs, intercurrent illness preventing further administration, investigator decision to withdraw therapy, participant withdrawal, pregnancy, noncompliance, or administrative reasons

Pembrolizumab 200 mg IV every 3 weeks for 2 years or until documented confirmed disease progression, intolerable toxicity, or patient or investigator decision to withdraw43

Pembrolizumab 200 mg every 3 weeks for 35 cycles or disease progression, unacceptable AEs, intercurrent illness preventing further administration, investigator decision to withdraw therapy, participant withdrawal, pregnancy, or administrative reasons

Comparator(s)

None

None

BV 1.8 mg/kg (maximum dose of 180 mg) every 3 weeks for 35 cycles or disease progression, unacceptable AEs, intercurrent illness preventing further administration, investigator decision to withdraw therapy, participant withdrawal, pregnancy, or administrative reasons

Duration

Phase

   Screening

28 days

28 days

28 days

   Treatment

Up to 35 doses, discontinuation, or event

Up to 2 years, discontinuation, or event

Up to 35 study doses, discontinuation, or event

   Follow-up

Occurs every 12 weeks after treatment discontinuation

Occurs every 12 weeks after treatment discontinuation

Occurs every 12 weeks after treatment discontinuation

Outcomes

Primary end point

  • Objective response rate

  • Safety

  • Objective response rate

  • Safety

  • Progression-free survival

  • Overall survival

  • Secondary and exploratory end points

  • Duration of response

  • Progression-free survival

  • Overall survival

  • Objective response rate using the Lugano classification

  • Progression-free survival

  • Duration of response

  • Overall survival

  • Progression-free survival (secondary definition)

  • Objective response rate

  • Complete remission rate

  • Safety

AE = adverse event; ASCT = autologous stem cell transplant; BV = brentuximab vedotin; cHL = classical Hodgkin lymphoma; CR = complete response; ECOG = Eastern Cooperative Oncology Group; PR = partial response.

Source: Study protocols for the KEYNOTE-051, KEYNOTE-087, and KEYNOTE-204 studies and Chen et al. (2019).

Description of Studies

KEYNOTE-051

The KEYNOTE-05132 study was a nonrandomized, open-label, single-arm trial of pembrolizumab 2 mg/kg administered every 3 weeks in 7 pediatric patients with cHL aged 3 years to 18 years. A 28-day screening period was performed before patient enrolment to collect necessary laboratory, diagnostic, and demographic information and assess study eligibility. The KEYNOTE-051 study evaluated safety and efficacy including ORR, DOR, PFS, and OS for 35 cycles of treatment or until discontinuation due to disease progression or AE. Post-treatment follow-up assessments occurred every 12 weeks. The study was funded by the sponsor and had a data cut-off date of January 2020.

KEYNOTE-087

The KEYNOTE-08733 study was a nonrandomized, single-arm study of pembrolizumab 200 mg administered every 3 weeks in adult patients with cHL. A 28-day screening period was performed before patient enrolment to collect necessary laboratory, diagnostic, and demographic information and assess study eligibility. The study evaluated ORR, PFS, DOR, HRQoL, and OS with a treatment duration up to 2 years, or until discontinuation of treatment due to disease progression, or occurrence of AE. Post-treatment follow-up assessments occurred every 12 weeks. The study was funded by the sponsor with a data cut-off date of March 2019. The study consisted of 3 cohorts:

KEYNOTE-204

The KEYNOTE-20434 study was a phase III, randomized (1:1 ratio), active controlled, open-label clinical trial comparing pembrolizumab 200 mg administered intravenously every 3 weeks (N = 151) with BV 1.8 mg/kg (maximum dose of 180 mg) administered intravenously every 3 weeks (N = 153) in adult patients with cHL. A 28-day screening period was performed before patient enrolment to collect necessary laboratory, diagnostic, and demographic information and assess study eligibility. The study evaluated PFS, OS, ORR, DOR, time to response, HRQoL, and safety for 35 cycles of treatment or until early discontinuation due to disease progression, unacceptable AEs, or other reasons to withdraw therapy. Post-treatment follow-up assessments occurred every 12 weeks. The study was funded by the sponsor with data cut-off date of February 2020. A diagram of the KEYNOTE-204 study design is provided in Figure 2.

Inclusion and Exclusion Criteria

The KEYNOTE-051 study recruited children and adolescents aged 3 years to 18 years with cHL who were either refractory to front-line therapy, high-risk and relapsed from front-line therapy, or relapsed or refractory to second-line therapy. The KEYNOTE-051 study did not explicitly recruit patients who failed ASCT or were ineligible for salvage chemotherapy and ASCT. The KEYNOTE-051 study recruited those with a Lansky Play-Performance Scale score of 50 or greater for children 16 years and younger or a Karnofsky score of 50 or greater in children 16 years and older. The KEYNOTE-051 study also excluded those who received prior systemic anticancer therapy including investigational agents within 2 weeks of the study’s start date or patients who had not recovered from AEs due to a previously administered agent. The KEYNOTE-087 and KEYNOTE-204 studies recruited adults 18 years or older with an ECOG score of 0 or 1. The KEYNOTE-087 study divided patients into 3 cohorts as follows.

Finally, patients recruited into the KEYNOTE-204 study had relapsed or refractory cHL and met 1 of the following criteria.

Both the KEYNOTE-087 and KEYNOTE-204 studies excluded those who received a prior monoclonal antibody within 4 weeks before the study’s start date or who had not recovered from AEs due to agents administered more than 4 weeks earlier. Both studies also excluded those who had prior chemotherapy, targeted small molecule therapy, or radiation therapy within 2 weeks before the study’s start date or who had not recovered from AEs due to a previously administered agent.

Figure 2: Study Design of KEYNOTE-204

Study design of the KEYNOTE-204 trial, demonstrating patient movement from screening (300 patients enrolled) to randomized treatment with pembrolizumab (n = 150) or brentuximab vedotin (n = 150), and to the follow-up period.

Source: Clinical Study Reports for KEYNOTE-204

Baseline Characteristics

Patients in the KEYNOTE-051 study had a median age of 15 years while the median age in the KEYNOTE-087 and KEYNOTE-204 studies ranged from 32.0 years to 40.0 years. The proportion of female patients ranged from 41.2% among BV patients in the KEYNOTE-204 study to 47.8% among cohort 1 of the KEYNOTE-087 study. The proportion of patients with an ECOG score of 0 ranged from 42.0% in cohort 1 of KEYNOTE-087 to 65.4% among BV patients from the KEYNOTE-204 study. The proportion of patients with an ECOG score of 0 was 54.3% and 48.3% in cohorts 2 and 3, respectively, of KEYNOTE-087, and 57.0% in the pembrolizumab arm of the KEYNOTE-204 study. Cohorts 1 and 3 of the KEYNOTE-087 study had higher rates of prior radiation use (46.4% and 40.0%, respectively) relative to either arm in the KEYNOTE-204 study (pembrolizumab: 38.4% and BV: 39.9%), while those in cohort 2 had lower rates (25.9%). Patients in either arm of the KEYNOTE-204 study had more bulky disease (pembrolizumab: 23.2% and BV: 16.3%) relative to any cohort in the KEYNOTE-087 study (cohort 1: 2.9%, cohort 2: 6.2%, and cohort 3: 1.7%). Baseline B symptoms were present in 30.4%, 33.3%, and 31.7% of patients in cohort 1, cohort 2, and cohort 3 of the KEYNOTE-087 study, respectively. Baseline B symptoms were also present in 28.5% and 23.5% of pembrolizumab and BV patients, respectively, in the KEYNOTE-204 study. The 2 arms within the KEYNOTE-204 study seem relatively balanced except that pembrolizumab patients had higher rates of bulky disease (23.2% versus 16.3%). A complete summary of baseline characteristics is provided in Table 18. Patients in the KEYNOTE-204 study were permitted to be treated with a subsequent anticancer medication after pembrolizumab or BV was discontinued. Almost all patients randomized to BV (97.4%) received a subsequent anticancer therapy while 70.2% of pembrolizumab-treated patients did so. Those randomized to BV were more likely to cross over and subsequently receive pembrolizumab (17.8% versus 1.4% of patients originally randomized to pembrolizumab and retreated with pembrolizumab). Those originally randomized to BV were also more likely to receive nivolumab (19.7%) relative to those randomized to pembrolizumab (3.4%). Finally, 25.0% of patients originally randomized to pembrolizumab received BV while 4.6% of patients originally randomized to BV were retreated with BV (Table 19 and Appendix 2).

Table 18: Summary of Baseline Characteristics (Intention-to-Treat Analysis)

Characteristic

KEYNOTE-051

KEYNOTE-087

KEYNOTE-204

Pembrolizumab

N = 7

Cohort 1

N = 69

Cohort 2

N = 81

Cohort 3

N = 60

Pembrolizumab

N = 151

BV

N = 153

Median age, years (range)

15.0

(12 to 17)

34.0

(19 to 64)

40.0

(20 to 76)

32.0

(18 to 73)

36.0

(18.0 to 84.0)

35.0

(18.0 to 83.0)

Mean age, years (SD)

15.0 (1.7)

37.0 (10.9)

42.3 (17.4)

36.8 (13.4)

41.9 (17.5)

40.8 (17.1)

Female, n (%)

3 (42.9)

33 (47.8)

38 (46.9)

26 (43.3)

67 (44.4)

63 (41.2)

Male, n (%)

4 (57.1)

36 (52.2)

43 (53.1)

34 (56.7)

84 (55.6)

90 (58.8)

Race, n (%)

   American Indian/

   Alaska Native

0 (0.0)

0 (0.0)

1 (1.2)

0 (0.0)

1 (0.7)

0 (0.0)

   Asian

0 (0.0)

7 (10.1)

4 (4.9)

1 (1.7)

13 (8.6)

13 (8.5)

   Black

0 (0.0)

2 (2.9)

2 (2.5)

3 (5.0)

4 (2.6)

8 (5.2)

   Multiracial

1 (14.3)

2 (2.9)

0 (0.0)

0 (0.0)

4 (2.6)

5 (3.3)

   Native Hawaiian/

   Pacific Islander

0 (0.0)

NR

NR

NR

1 (0.7)

0 (0.0)

   White

6 (85.7)

57 (82.6)

73 (90.1)

55 (91.7)

119 (78.8)

115 (75.2)

   Missing

0 (0.0)

1 (1.4)

1 (1.2)

0 (1.7)

9 (6.0)

12 (7.8)

Ethnicity, n (%)

   Hispanic or Latino

3 (42.9)

7 (10.1)

5 (6.2)

3 (5.0)

24 (15.9)

20 (13.1)

   Not Hispanic or Latino

4 (57.1)

43 (62.3)

63 (77.8)

48 (80.0)

111 (73.5)

115 (75.2)

   NR

0 (0.0)

9 (13.0)

9 (11.1)

4 (6.7)

8 (5.3)

10 (6.5)

   Unknown

0 (0.0)

10 (14.5)

4 (4.9)

5 (8.3)

6 (4.0)

5 (3.3)

Region of residence, n (%)

   North America

NR

13 (18.8)

20 (24.7)

18 (30.0)

27 (17.9)

30 (19.6)

   Europe

NR

NR

NR

NR

49 (32.5)

46 (30.1)

   Japan

NR

NR

NR

NR

9 (6.0)

7 (4.6)

   Rest of world

NR

56 (81.2)

61 (75.3)

42 (70.0)

66 (43.7)

70 (45.8)

Disease subtype, n (%)

   Mixed cellularity

NR

9 (13.0)

10 (12.3)

5 (8.3)

23 (15.2)

17 (11.1)

   Nodular sclerosis

NR

55 (79.7)

65 (80.2)

49 (81.7)

119 (78.8)

127 (83.0)

   Lymphocyte depleted

NR

0 (0.0)

4 (4.9)

1 (1.7)

3 (2.0)

3 (2.0)

   Lymphocyte rich

NR

4 (5.8)

1 (1.2)

3 (5.0)

1 (0.7)

1 (0.7)

   Missing

NR

1 (1.4)

1 (1.2)

2 (3.3)

5 (3.3)

5 (3.3)

ECOG Performance, n (%)

   0

NR

29 (42.0)

44 (54.3)

29 (48.3)

   1

NR

39 (56.5)

37 (45.7)

31 (51.7)

   2

NR

1 (1.4)

0 (0.0)

0 (0.0)

Prior ASCT, n (%)

   Yes

NR

NR

56 (37.1)

56 (36.6)

   No

NR

NR

95 (62.9)

97 (63.4)

Disease status after front-line therapy, n (%)

   Primary

NR

NR

61 (40.4)

62 (40.5)

   Relapsed < 12 months

NR

NR

42 (27.8)

42 (27.5)

   Relapsed ≥ 12 months

NR

NR

48 (31.8)

49 (32.0)

Refractory or relapsed after any life of therapy, n (%)

   Yes

NR

NR

149 (98.7)

153 (100.0)

   No

NR

NR

2 (1.3)

0 (0.0)

Response to first regimen, n (%)

   Refractory

NR

NR

47 (31.1)

40 (26.1)

   Relapse

NR

NR

97 (64.2)

102 (66.7)

   Other

NR

NR

7 (4.6)

11 (7.2)

Response to previous regimen, n (%)

   Refractory

NR

NR

65 (43.0)

64 (41.8)

   Untreated relapse

NR

NR

50 (33.1)

61 (39.9)

   Other

NR

NR

36 (23.8)

28 (18.3)

Number of lines of prior therapy

   Mean (SD)

NR

4.6 (1.7)

4.0 (1.6)

3.5 (1.8)

   Median (range)

NR

4.0

(2 to 12)

4.0

(1 to 11)

3.0

(2 to 10)

2.0

(1 to 10)

3.0

(1 to 11)

Refractory or relapsed after ≥ 3 lines of therapy, n (%)

   Yes

NR

69 (100.0)

81 (100.0)

60 (100.0)

NR

   No

NR

0 (0.0)

0 (0.0)

0 (0.0)

NR

Time of relapse since ASCT failure, n (%)

   ≥ 12 months

NR

37 (53.6)

NR

7 (11.7)

NR

   < 12 months

NR

32 (46.4)

NR

53 (88.3)

NR

   Missing

NR

0 (0.0)

NR

0 (0.0)

NR

   Mean, months (SD)

NR

30.4 (40.0)

NR

6.3 (11.8)

NR

   Median, months (range)

NR

12.6

(2.5 to 247.9)

NR

1.9

(0.4 to 76.0)

NR

PD-L1 status, n (%)

   ≥ 1%

NR

NR

142 (94.0)

133 (86.9)

   < 1%

NR

NR

0 (0.0)

3 (2.0)

   Missing

NR

NR

9 (6.0)

17 (11.1)

Prior use of BV, n (%)

   Yes

NR

69 (100.0)

81 (100.0)

25 (41.7)

5 (3.3)

10 (6.5)

   No

NR

0 (0.0)

0 (0.0)

35 (58.3)

146 (96.7)

143 (93.5)

Prior radiation, n (%)

   Yes

NR

32 (46.4)

21 (25.9)

24 (40.0)

58 (38.4)

61 (39.9)

   No

NR

37 (53.6)

60 (74.1)

36 (60.0)

93 (61.6)

92 (60.1)

Bulky disease, n (%)

   Yes

NR

2 (2.9)

5 (6.2)

1 (1.7)

35 (23.2)

25 (16.3)

   No

NR

67 (97.1)

76 (93.8)

59 (98.3)

116 (76.8)

128 (83.7)

Baseline B symptoms, n (%)

   Yes

NR

21 (30.4)

27 (33.3)

19 (31.7)

43 (28.5)

36 (23.5)

   No

NR

48 (69.6)

54 (66.7)

41 (68.3)

108 (71.5)

116 (75.8)

   Missing

NR

0 (0.0)

0 (0.0)

0 (0.0)

0 (0.0)

1 (0.7)

Baseline bone marrow involvement, n (%)

   Yes

NR

3 (4.3)

5 (6.2)

3 (5.0)

12 (7.9)

5 (3.3)

   No

NR

66 (95.7)

75 (92.6)

57 (95.0)

139 (92.1)

148 (96.7)

   Missing

NR

0 (0.0)

1 (1.2)

0 (0.0)

0 (0.0)

0 (0.0)

ASCT = autologous stem cell transplant; BV = brentuximab vedotin; ECOG = Eastern Cooperative Oncology Group; NR = not reported; SD = standard deviation.

Source: Clinical Study Reports for KEYNOTE-051, KEYNOTE-087, KEYNOTE-204, and Common Technical Document Section 2.7.3.

Interventions

KEYNOTE-051

KEYNOTE-051 studied pembrolizumab 2 mg/kg intravenously every 3 weeks until 35 cycles were administered, disease progression, unacceptable AEs, intercurrent illness preventing further administration, investigator decision to withdraw therapy, patient withdrawal, pregnancy, noncompliance, or administrative reasons. Patients were prohibited from concurrently using granulocyte-macrophage colony-stimulating factor, immunotherapy (other than pembrolizumab), chemotherapy, biologic therapy, investigational agents other than pembrolizumab, radiation, live vaccines, and glucocorticoids for any reason other than to treat an AE. Otherwise, patients were permitted to concurrently receive any medication necessary for the patient’s welfare so long as they adhered to standards of medical care and medication use was documented.

KEYNOTE-087

In each of the 3 cohorts of the KEYNOTE-087 study, pembrolizumab 200 mg was administered intravenously every 3 weeks for 2 years or until documented and confirmed disease progression, intolerable toxicity, or patient or investigator decision to withdraw. Patients were prohibited from concurrently using granulocyte-macrophage colony-stimulating factor, immunotherapy (other than pembrolizumab), chemotherapy, biologic therapy, investigational agents other than pembrolizumab, radiation, live vaccines, and glucocorticoids for any reason other than to treat an AE. Otherwise, patients were permitted to concurrently receive any medication necessary for the patient’s welfare so long as they adhered to standards of medical care and medication use was documented.

KEYNOTE-204

Pembrolizumab 200 mg or BV 1.8 mg/kg (maximum dose of 180 mg) was administered intravenously every 3 weeks for 35 cycles or until disease progression, unacceptable AEs, intercurrent illness preventing further administration, investigator decision to withdraw therapy, patient withdrawal, pregnancy, or administrative reasons. Patients in either arm were permitted to receive subsequent anticancer therapy after discontinuing pembrolizumab or BV. Further, those originally randomized to pembrolizumab were able to then receive BV while those originally randomized to BV were permitted to receive pembrolizumab. Patients were prohibited from concurrently using granulocyte-macrophage colony-stimulating factor, immunotherapy (other than pembrolizumab), chemotherapy (other than BV), biologic therapy, investigational agents other than pembrolizumab and BV, radiation, live vaccines, glucocorticoids for any reason other than to treat an AE, and in those receiving BV, potent CYP3A4 inhibitors or inducers or P-glycoprotein inhibitors. Otherwise, patients were permitted to concurrently receive any medication necessary for the patient’s welfare so long as they adhered to standards of medical care and medication use was documented.

In all 3 trials, if a patient experienced an immune-mediated AE, they could be treated with corticosteroids, anti-inflammatory agents if symptoms did not improve following corticosteroid treatment, insulin, non-selective beta blockers, thyroid hormone replacement therapy, antihistamines, nonsteroidal anti-inflammatory drugs, acetaminophen, opioids, vasopressors, and epinephrine. Supportive care was permitted as deemed necessary by the treating physician.

Table 19: Summary of Subsequent Use of Anticancer Medication Utilization in KEYNOTE-204; Greater Than 5% Utilization in Either Arm

Anticancer therapy, n (%)

Pembrolizumab

N = 148

Brentuximab vedotin

N = 152

Total

104 (70.2)

148 (97.4)

Bendamustine and brentuximab vedotin

9 (6.1)

6 (3.9)

Brentuximab vedotin

37 (25.0)

7 (4.6)

Nivolumab

5 (3.4)

30 (19.7)

Pembrolizumab

2 (1.4)

27 (17.8)

Source: Clinical Study Report for KEYNOTE-204.

Outcomes

A list of efficacy end points identified in the CADTH review protocol that were assessed in the clinical trials included in this review is provided in Table 20. These end points are further summarized below. A detailed discussion and critical appraisal of the outcome measures are provided in Appendix 3.

Table 20: Summary of Outcomes of Interest Identified in the CADTH Review Protocol

Outcome measure

KEYNOTE-051

KEYNOTE-087

KEYNOTE-204

Progression-free survival

Secondary

Secondary

Primary

Overall survival

Secondary

Secondary

Primary

Objective response rate

Primary

Primary

Secondary

Complete remission rate

Measured but not specified as primary, secondary, or exploratory

Secondary

Secondary

Duration of response

Secondary

Secondary

Exploratory

Time to response

Secondary

Secondary

Exploratory

Health-related quality of life

Not reported

Exploratory

Exploratory

Progression-Free Survival

In the KEYNOTE-051 and KEYNOTE-087 studies, PFS was defined as time from first dosing date to the first documented progressive disease, death due to any cause, or start of new anticancer medication, whichever came first by blinded independent central radiology assessment and using International Working Group (IWG) response criteria. The IWG guidelines utilize diagnostic imaging, immunohistochemistry, and flow cytometry to define response to treatment in non-Hodgkin and HL.44

In the KEYNOTE-204 study, PFS was assessed by blinded independent central review according to the IWG response criteria including clinical and imaging data following autologous or allogenic stem cell transplant. PFS was defined as the time from randomization to the first documentation of progression or death from any cause. Patients were censored at the last disease assessment if they received an ASCT or another anticancer therapy. These analyses considered ASCT or initiation of another anticancer treatment as a censoring event. A sensitivity analysis of PFS considered the use of another anticancer medication as a progression event but otherwise had the same definition of PFS.

Overall Survival

In the KEYNOTE-051 and KEYNOTE-087 studies, OS was defined as time from first dose to the date of death. In KEYNOTE-204, OS was defined as the time from randomization to death from any cause. In the KEYNOTE-204 study, patients without death were censored at the date of the last assessment.

Objective Response Rate

In the KEYNOTE-051, KEYNOTE-087, and KEYNOTE-204 studies, ORR was defined as the proportion of patients who had a complete or partial response. All studies assessed response by blinded independent central review and used the IWG criteria.

Complete Remission Rate

Like ORR, complete remission rate was assessed by blinded independent central review using the IWG criteria in the KEYNOTE-051, KEYNOTE-087, and KEYNOTE-204 studies.

Duration of Response

In the KEYNOTE-051 and KEYNOTE-087 studies, DOR was defined as the time from first response to documented progressive disease or death from any cause in patients who achieved a partial response or better using IWG response criteria and by blinded independent central review. Those without a response were excluded from this analysis. DOR in the KEYNOTE-204 study was also assessed by blinded independent central review using IWG criteria but a clear definition of DOR was not provided.

Time to Response

No information was provided regarding time to response in the KEYNOTE-051, KEYNOTE-087, or KEYNOTE-204 studies.

Health-Related Quality of Life

HRQoL was not measured in the KEYNOTE-051 study. In the KEYNOTE-087 and KEYNOTE-204 studies, HRQoL was measured by the EORTC QLQ-C30 and EQ-5D-3L. The EORTC QLQ-30 is a widely used, cancer-specific HRQoL instrument consisting of 30 items measuring 5 functional dimensions (physical, role, cognitive, emotional, and social), 3 symptoms dimensions (fatigue, nausea/vomiting, and pain), 6 additional items (dyspnea, sleep disturbance, appetite loss, constipation, diarrhea, and financial impact), and a global HRQoL measure. The minimal important difference for adult cancer patients on the EORTC QLQ-C30 scale is 5 but no minimal important difference was identified specifically for patients with cHL.45 EQ-5D-3L is another standard instrument to measure health outcomes and is particularly useful to develop economic models. EQ-5D-3L measures mobility, self-care, usual activities, pain/discomfort, and anxiety/depression using a 3-point ordinal scale. These measurements can be pooled into a single utility score. Further, the EQ-5D-3L contains a visual analogue scale ranging from 0 to 100 so that participants may rate their general health state. Each of the HRQoL questionnaires were conducted at baseline and at 24 weeks so that the change could be calculated and compared between groups. The minimal important difference for adult cancer patients on the EQ-5D-3L visual analogue scale is 6 to 10 and the minimal important difference for American patients with cancer on the EQ-5D-3L utility scale is 0.05 to 0.08.46 No minimal important difference on the visual analogue scale or utility portion of the EQ-5D-3L scale for patients with cHL was identified.

Harms

All 3 studies assessed AEs, serious AEs, and immune-mediated AEs. An AE was defined as any untoward medical occurrence in a patient which does not necessarily have a causal relationship with the treatment. Serious AEs were defined as those which result in death, are life threatening, result in persistent or significant disability/incapacity, result in or prolong an existing inpatient hospitalization, result in a congenital anomaly/birth defect, is another important medical event, results in the development of a new cancer (different from the cancer under investigation), or is associated with an overdose. Immune-mediated AEs were defined as AEs of unknown etiology associated with drug exposure and consistent with an immune phenomenon. The following are examples: pneumonitis, diarrhea/colitis, elevated aspartate aminotransferase, elevated alanine aminotransferase, elevated bilirubin, type 1 diabetes mellitus, hypophysitis, hyperthyroidism, hypothyroidism, nephritis, renal dysfunction, and myocarditis.

Statistical Analysis

Progression-Free Survival

In KEYNOTE-087, PFS was estimated using the Kaplan-Meier method. In the KEYNOTE-204 study, PFS was analyzed using a stratified log rank test and the Kaplan-Meier method and the hazard ratio was estimated using a stratified Cox regression model using the Efron method to handle ties. The analysis was stratified by prior ASCT and cHL status after front-line therapy (primary refractory, relapsed disease < 12 months after completion of first-line therapy, or relapse 12 months or longer after completing first-line therapy). In the KEYNOTE-204 study, the analysis considered ASCT or initiation of another anticancer treatment as a censoring event. A sensitivity analysis of PFS considered the use of another anticancer medication as a progression event but otherwise had the same definition of PFS.

Overall Survival

In the KEYNOTE-087 study, OS was estimated using the Kaplan-Meier method. In the KEYNOTE-204 study, OS was analyzed in the same manner as the PFS analysis. The study protocol stated that if required to adjust for patients receiving subsequent anticancer therapies (following pembrolizumab or BV) in the OS analysis, the rank-preserving structural failure time and 2-stage analysis methods would be used. Rank-preserving structural failure time assumes all patients would receive equal benefit from identical interventions. This method compares time on and off treatment to estimate survival times without treatment and a treatment effect adjusted for subsequent utilization of anticancer therapy. The 2-stage adjustment assumes subsequent utilization of anticancer therapy only occurs following disease progression and uses this point to establish a “secondary” baseline. Within the control group, the treatment effect is estimated between those who do and do not subsequently use anticancer therapy that adjusts for “secondary” baseline characteristics. The incremental treatment effect between these groups is then used to discount the treatment effect observed in those who subsequently use additional anticancer therapies, which are then compared with the experimental group to estimate the treatment effect adjusted for subsequent use of anticancer therapies.

Objective Response Rate

In the KEYNOTE-087 and KEYNOTE-204 studies, ORR was estimated by a point estimate and 95% 2-sided binomial exact CIs using the Clopper-Pearson method. In the KEYNOTE-204 study, the difference in ORR was analyzed using the Miettinen and Nurminen method, weighted by stratum. In this analysis, data were stratified by previous ASCT and cHL status following front-line therapy (primary refractory, relapsed disease < 12 months after completion of first-line therapy, or relapse 12 months or longer after completing first-line therapy). Patients with missing data were assumed to be non-responders.

Complete Response Rate

In the KEYNOTE-087 study, complete response rate analysis consisted of the point estimate and 95% 2-sided exact CI. In the KEYNOTE-204 study, complete response rate was analyzed as in the ORR analysis.

Duration of Response

In the KEYNOTE-087 study, DOR was estimated using the Kaplan-Meier method. Patients without progression were censored on the date of the most recent assessment. In the KEYNOTE-204 study, no explicit statistical analysis was outlined for DOR.

Time to Response

In the KEYNOTE-087 and 204 studies, no explicit statistical analysis was outlined for time to response.

Health-Related Quality of Life

In the KEYNOTE-087 study, HRQoL data were collected at baseline and 24 weeks later in all individuals. The difference in scores between baseline and week 24 was analyzed using a longitudinal data analysis model adjusting for time and ECOG status and then estimated using a least squares mean score and standard error. In the KEYNOTE-204 study, no explicit statistical analysis was outlined for HRQoL.

Harms

In the KEYNOTE-087 study, only descriptive statistics were provided. In the KEYNOTE-204 study, descriptive statistics including point estimates and 95% CIs were estimated and the unstratified Miettinen and Nurminen method was used to assess between-treatment differences.

Interim Analyses

The KEYNOTE-204 study results presented in this report constitute the second of 4 planned interim analyses. The study protocol planned for 1 interim analysis of PFS and 2 interim analyses of OS. An interim PFS analysis was to be conducted 3 months after all patients had been enrolled and once 110 PFS events had occurred. A final PFS analysis was planned once 221 events had occurred. If the PFS hypothesis was not rejected at the interim, then the first interim OS analysis would occur with the final PFS analysis. This assumes that 91 OS events would have been observed at this point. If the PFS hypothesis was rejected at the interim analysis then the interim OS analysis would occur 1 year from the interim PFS analysis or when 91 OS events occurred, whichever came first. If the OS hypothesis was not rejected at that point, a second interim OS analysis would occur at 119 events and if the OS hypothesis was not rejected at that point, a final OS analysis was planned to occur at 146 events. No interim analysis was planned for ORR. The final ORR analysis will occur with the final PFS analysis (when 221 PFS events have occurred).

Multiplicity

In the KEYNOTE-087 study, no multiplicity adjustments were required as each cohort was evaluated independently. All tests of significance were controlled at a 1-sided alpha of 0.025. In the KEYNOTE-204 study, the Mauer and Bretz method was used to allocate and re-allocate type I error between hypotheses and group sequential methods to allocate alpha between interim and final analyses. If the null hypothesis was rejected, the type I error allocated to that hypothesis would be redistributed to other hypotheses. For example, multiplicity could be controlled at 2.5% (1-sided) with 1.25% originally allocated to PFS and OS and none allocated to the ORR hypothesis. If the PFS null hypothesis was rejected, then 0.625% would be allocated to both the OS and ORR test. If the ORR null hypothesis was rejected, then 0.625% would be allocated to the OS analysis. OS testing was planned to occur at the 1.25% level if the PFS null hypothesis was not rejected, at 1.875% if the PFS but not ORR null hypotheses were rejected, or 2.5% if both the PFS and ORR null hypotheses were rejected.

Power Calculations

In the KEYNOTE-087 study, for cohorts 1 and 3, there is 93% power, at a 1-sided 2.5% alpha, to detect a 35% or higher ORR between pembrolizumab and a fixed control rate of 15% using the exact binomial test. This would require at least 16 responses if 60 patients are recruited. In cohort 2, there is 93% power, at a 1-sided 2.5% alpha, to detect a 20% or higher overall response rate in the pembrolizumab arm versus the fixed control rate of 5% using exact binomial test. This would require at least 8 responses out of 60 patients.

On the basis of 194 PFS events, the study had an 85% power to detect a hazard ratio of 0.622 (pembrolizumab versus BV) at and alpha of 1.2% (1-sided), assuming PFS would follow an exponential distribution with a median of 5·6 months in the control.

Table 21: Statistical Analysis of Efficacy End Points

End point

Statistical model

Adjustment factors

Sensitivity analyses

KEYNOTE-051

PFS

Kaplan-Meier

None

None

OS

Kaplan-Meier

None

None

ORR

Truncated sequential probability ratio test

None

None

CR

Not reported

Not reported

Not reported

DOR

Kaplan-Meier

None

None

Time to response

Not reported

Not reported

Not reported

KEYNOTE-087

PFS

Kaplan-Meier

None

None

OS

Kaplan-Meier

None

None

ORR

None

None

None

CR

Not reported

Not reported

Not reported

DOR

Kaplan-Meier

None

None

Time to response

Not reported

Not reported

Not reported

HRQoL

Longitudinal analysis model

Time, ECOG status

None

KEYNOTE-204

PFS and OS

Kaplan-Meier

ASCT and cHL status after front-line therapy

To assess censoring in PFS analysis

ORR and CR

Miettinen-Nurminen

ASCT and cHL status after front-line therapy

None

DOR

Not reported

Not reported

Not reported

Time to response

Not reported

Not reported

Not reported

HRQoL

Not reported

Not reported

Not reported

ASCT = autologous stem cell transplant; cHL = classical Hodgkin lymphoma; CR = complete response; DOR = duration of response; ECOG = Eastern Cooperative Oncology Group; HRQoL = health-related quality of life; ORR = objective response rate; OS = overall survival; PFS = progression-free survival.

Analysis Populations

In the KEYNOTE-051 and KEYNOTE-087 studies, all patients who received at least 1 dose of study medication were analyzed in the efficacy and safety analyses. In the KEYNOTE-204 study, the intention-to-treat population was used in the efficacy analysis and the safety analysis consisted of all patients who received at least 1 dose of a study medication.

Results

Patient Disposition

No patient in the KEYNOTE-051 study discontinued the trial. Of the 7 patients in the KEYNOTE-051 study, 2 are still receiving treatment while 5 others had discontinued therapy at the time of data cut-off. In the KEYNOTE-087 study, 72.5%, 84.0%, and 76.7% of patients discontinued treatment in cohorts 1, 2, and 3, respectively, at the time of data cut-off. In the KEYNOTE-204 study, 338 patients were screened, 151 patients were randomized to pembrolizumab, and 153 were randomized to BV. Fewer patients in the pembrolizumab arm (13.2%) discontinued the trial compared to those in the BV arm (28.1%). A notable difference in trial discontinuation was due to deaths (10.6% in the pembrolizumab arm and 17.6% in the BV arm) and withdrawals (2.0% in the pembrolizumab arm and 8.5% in the BV arm). Similarly, treatment discontinuations were lower in the pembrolizumab arm (74.3%) than in the BV arm (96.1%). Notable differences in the reasons for treatment discontinuations were due to AEs (pembrolizumab: 13.5% versus BV: 19.1%) and progressive disease (pembrolizumab: 39.2% versus BV: 49.3%). Full details regarding patient disposition are available in Table 22.

Table 22: Patient Disposition

Disposition

KEYNOTE-051

KEYNOTE-087

KEYNOTE-204

Pembrolizumab

Cohort 1

Cohort 2

Cohort 3

Pembrolizumab

Brentuximab vedotin

Screened, n

7

69

81

60

338

Randomized

NA

NA

151

153

Ongoing in trial

Discontinued from trial

   Death

   Lost to follow-up

   Physician decision

   Withdrawal by subject/family

7 (100)

0 (0)

0 (0)

0 (0)

0 (0)

0 (0)

NR

131 (86.8)

20 (13.2)

16 (10.6)

0 (0.0)

1 (0.7)

3 (2.0)

110 (71.9)

43 (28.1)

27 (17.6)

3 (2.0)

0 (0.0)

13 (8.5)

Treated

7 (100)

69 (100)

81 (100)

60 (100)

148 (98.0)

152 (99.3)

Completed treatment

Treatment ongoing

Discontinued treatment

   Adverse event

   Bone marrow transplant

   Clinical progression

   Complete response

   Excluded medication

   Noncompliance

   Non-study anticancer therapy

   Physician decision

   Progressive disease

   Protocol deviation

   Withdrawal by subject/family

   Lost to follow-up

   Pregnancy

0 (0.0)

2 (28.6)

5 (71.4)

1 (14.3)

0 (0.0)

0 (0.0)

0 (0.0)

0 (0.0)

0 (0.0)

1 (14.3)

1 (14.3)

2 (28.6)

0 (0.0)

0 (0.0)

0 (0.0)

0 (0.0)

19 (27.5)

NR

50 (72.5)

8 (11.6)

1 (1.4)

3 (4.3)

8 (11.6)

NR

NR

NR

3 (4.3)

23 (33.3)

NR

3 (4.3)

1 (1.4)

0 (0.0)

13 (16.0)

NR

68 (84.0)

5 (6.2)

2 (2.5)

1 (1.2)

9 (11.1)

NR

NR

NR

6 (7.4)

37 (45.7)

NR

5 (6.2)

2 (2.5)

1 (1.2)

14 (23.3)

NR

46 (76.7)

5 (8.3)

1 (1.7)

1 (1.7)

11 (18.3)

NR

NR

NR

1 (1.7)

26 (43.3)

NR

1 (1.7)

0 (0.0)

0 (0.0)

25 (16.9)

13 (8.8)

110 (74.3)

20 (13.5)

16 (10.8)

1 (0.7)

1 (0.7)

0 (0.0)

1 (0.7)

6 (4.1)

4 (2.7)

58 (39.2)

1 (0.7)

2 (1.4)

NR

NR

3 (2.0)

3 (2.0)

146 (96.1)

29 (19.1)

17 (11.2)

6 (3.9)

3 (2.0)

1 (0.7)

0 (0.0)

4 (2.6)

6 (3.9)

75 (49.3)

1 (0.7)

4 (2.6)

NR

NR

Median duration of exposure, days (range)

NR

506.0

(1 to 1,261)

254.0

(1 to 1,250)

399.5

(66 to 1,224)

305.0

(1 to 814)

146.5

(1 to 794)

ITT, n

7

69

81

60

151

153

Safety, n

7

69

81

60

148

152

ITT = intention to treat; NA = not applicable; NR = not reported.

Note: Values are n (%) unless otherwise indicated.

Source: Clinical Study Reports for the KEYNOTE-051, KEYNOTE-087, and KEYNOTE-204 studies.

Exposure to Study Treatments

In the KEYNOTE-051 study the median exposure to pembrolizumab was 344 days. Those in the KEYNOTE-087 study were exposed to pembrolizumab for a median of 506, 254, and 399.5 days in cohorts 1, 2, and 3, respectively. In the KEYNOTE-204 study, the median duration of exposure to pembrolizumab was 305 days and the median duration of exposure to BV was 146.5 days.

Efficacy

Only the efficacy outcomes that were identified in the review protocol are summarized below and in Table 23.

Progression-Free Survival

In the KEYNOTE-051 study, 3 patients (42.9%) experienced an event (disease progression or death). In the KEYNOTE-087 study, there were 43 (62.3%), 54 (66.7%), and 36 (60.0%) events in cohorts 1, 2, and 3, respectively. In the KEYNOTE-204 study, the proportion of patients experiencing an event were similar between the pembrolizumab (53.6%) and BV (57.5%) arms. In the KEYNOTE-051 study, the median PFS was reported to be 11.1 months (95% CI, 2.6 to not reported). In the KEYNOTE-087 study, median survival was reported to be 16.4 months (95% CI, 11.3 to 27.6), 11.1 months (95% CI, 7.3 to 13.5), and 19.4 (95% CI, 8.4 to 22.1) months in cohorts 1, 2, and 3, respectively. In the KEYNOTE-204 study, the median PFS was higher in the pembrolizumab arm (13.2 months; 95% CI, 10.9 to 19.4) than the BV arm (8.3 months; 95% CI, 5.7 to 8.8). In the KEYNOTE-051 study, the PFS rate at 12 months was 27.8% (no 95% CI reported). In the KEYNOTE-087 study, the PFS rate at 12 months was 61.3%, 43.0%, and 53.9% in cohorts 1, 2, and 3, respectively (no 95% CI reported). In the KEYNOTE-204 study, the 12-month PFS rate was higher in the pembrolizumab arm (53.9%; 95% CI, 45.0 to 61.9) than the BV arm (35.6%; 95% CI, 26.9 to 44.4). In the KEYNOTE-087 study, the 24-month PFS rate was 41.6%, 21.9%, and 34.0% in cohorts 1, 2, and 3, respectively (no 95% CI reported). In the KEYNOTE-204 study, the 24-month PFS rate was 35.4% (95% CI, 26.2 to 44.6) in the pembrolizumab arm and 25.4% (95% CI, 17.1 to 34.5) in the BV arm. The hazard ratio for time to progression was 0.65 (95% CI, 0.48 to 0.88), which was statistically significant (P = 0.0027).

In the KEYNOTE-204 study, the primary PFS analysis considered initiation of subsequent anticancer therapy or ASCT as a censoring event. A sensitivity analysis was conducted which treated these events as a progression event instead. In this analysis, 103 (68.2%) and 119 (77.8%) events were observed in the pembrolizumab and BV arms, respectively. The median PFS was higher in the pembrolizumab arm (9.5 months; 95% CI, 8.2 to 12.7) than the BV arm (5.7 months; 95% CI, 5.6 to 8.3). The hazard ratio for time to progression was 0.62 (95% CI, 0.48 to 0.82).

Figure 3: Kaplan-Meier Estimates of PFS in KEYNOTE-204

Depicts Kaplan-Meier progression-free survival curves for MK3475 200 mg and brentuximab vedotin from 0 to 42 months of follow-up. Curves start to diverge from around month 5 with MK3475 200 mg above and brentuximab vedotin below. The curves remain separated until all subjects at risk are censored around month 37. Patients at risk in the 2 groups are shown for the duration.

MK-3475 = pembrolizumab.

Source: Clinical Study Reports for KEYNOTE-204 studies.

Overall Survival

In the KEYNOTE-051 study, minimal information regarding OS was provided. In the KEYNOTE-087 study, 15.9%, 16.0%, and 15.0% of patients in cohorts 1, 2, and 3, respectively, died. |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||||||||||||||||||||||||||||||||||||||||||||||| Median survival was not reported in the KEYNOTE-051 study and not reached in the KEYNOTE-087 or KEYNOTE-204 studies. In the KEYNOTE-051 study, 100% of patients were alive at 12 months. In the KEYNOTE-087 study, OS at 12 months was 95.7%, 96.2% and 96.6% in cohorts 1, 2, and 3, respectively (95% CI not reported). |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| At 24 months in the KEYNOTE-087 study, 92.6%, 91.0%, and 89.4% of patients were alive in cohorts 1, 2, and 3, respectively (95% CI not reported).  |  |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||| |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||

Figure 4: ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||

Figure has been redacted at the request of the sponsor.

Note: Figure 4 has been redacted at the request of the sponsor in accordance with the CADTH Drug Reimbursement Review Confidentiality Guidelines.

Objective Response Rate

In the KEYNOTE-051 study, 42.9% (95% CI, 9.9 to 81.6) of patients experienced a partial or complete response. In the KEYNOTE-087 study, 78.3% (95% CI, 66.7 to 87.3), 64.2% (95% CI, 52.8 to 74.6), and 71.7% (95% CI, 58.6 to 82.5) of patients experienced a partial or complete response in cohorts 1, 2, and 3, respectively. In the KEYNOTE-204 study, more partial or completes responses were observed in the pembrolizumab arm relative to the BV arm (65.6%; 95% CI, 57.4 to 73.1 versus 54.2; 95% CI, 46.0 to 62.3), which was associated with a statistically insignificant 11.3% (95% CI, 0.2 to 22.1) difference in favour of pembrolizumab.

Complete Response Rate

In the KEYNOTE-051 study, 28.6% of patients (95% CI, 3.7 to 71.0) experienced a complete response. In the KEYNOTE-087 study, 26.1% (95% CI, 16.3 to 38.1), 25.9 (95% CI, 16.8 to 36.9), and 31.7% (95% CI, 20.3 to 45.0) of patients in cohorts 1, 2, and 3, respectively, experienced a complete response. In the KEYNOTE-204 study, the complete response rate was comparable between the pembrolizumab (24.5%; 95% CI, 17.9 to 32.2) and BV arms (24.2; 95% CI, 17.6 to 31.8).

Duration of Response

In the KEYNOTE-051 study, median DOR was not reached. In the KEYNOTE-087 study, the median DOR in cohorts 1, 2, and 3 were 25.0 months (range = 0 to 36.1), 11.1 months (range = 0 to 35.9), and 16.8 months (range = 0 to 39.1), respectively. In the KEYNOTE-204 study, the median DOR was higher among patients in the pembrolizumab arm (20.7 months; range = 0 to 33.2) than in patients in the BV arm (13.8 months; range = 0 to 33.9).

Time to Response

Median time to response in the KEYNOTE-051 study was 2.6 months (range = 2.1 to 2.8). The median time to response in cohort 1, cohort 2, and cohort 3 of the KEYNOTE-087 study were 2.7 months (range = 2.1 to 12.9), 2.8 months (range = 2.2 to 11.0), and 2.8 months (range = 2.6 to 16.5), respectively. Finally, the median time to response in the pembrolizumab arm of the KEYNOTE-204 study was 2.8 months (range = 1.0 to 31.2) and also 2.8 months (range = 1.3 to 7.3) in the BV arm.

Health-Related Quality of Life

HRQoL data were only measured in the KEYNOTE-087 and KEYNOTE-204 studies. In the KEYNOTE-087 study, the least squares mean square change in EORTC QLQ-C30 global health status between week 24 and baseline was 11.8, 13.9 and 6.6, in cohorts 1, 2, and 3, respectively. No CIs were reported in the KEYNOTE-087 study. In the KEYNOTE-204 study, the least squares mean change in EORTC QLQ-C30 global health status between baseline and week 24 was 8.60 points (95% CI, 3.89 to 13.31) higher in the pembrolizumab arm versus the BV arm. Consistent results were reported for the EORTC QLQ-C30 physical functioning scale (6.24; 95% CI, 1.87 to 10.62), EQ-5D-3L utility score (0.09; 95% CI, 0.04 to 0.14), and EQ-5D-3L visual analogue scale (6.12; 95% CI, 1.91 to 10.34).

Table 23: Key Efficacy Results in KEYNOTE-051, KEYNOTE-087, and KEYNOTE-204 (Intention-to-Treat Analysis)

Result

KEYNOTE-051

KEYNOTE-087

KEYNOTE-204

Pembrolizumab

N = 7

Cohort 1

N = 69

Cohort 2

N = 81

Cohort 3

N = 60

Pembrolizumab

N = 151

Brentuximab vedotin

N = 153

Progression-free survival (primary analysis)

Number of events, n (%)

3 (42.9)

43 (62.3)

54 (66.7)

36 (60.0)

81 (53.6)

88 (57.5)

Median survival, months (95% CI)

11.2

(2.6 to NR)

16.4

(11.3 to 27.6)

11.1

(7.3 to 13.5)

19.4

(8.4 to 22.1)

13.2

(10.9 to 19.4)

8.3

(5.7 to 8.8)

PFS at 12 months, % (95% CI)

27.8 (NR)

61.3 (NR)

43.0 (NR)

53.9 (NR)

53.9

(45.0 to 61.9)

35.6

(26.9 to 44.4)

PFS at 24 months, % (95% CI)

NR

41.6 (NR)

21.9 (NR)

34.0 (NR)

35.4

(26.2 to 44.6)

25.4

(17.1 to 34.5)

Hazard ratio (95% CI)

NA

NA

0.65 (0.48 to 0.88); P = 0.00271a

Progression-free survival (sensitivity analysis)

Number of events, n (%)

NA

NA

103 (68.2)

119 (77.8)

Median, months (95% CI)

NA

NA

9.5 (8.2 to 12.7)

5.7 (5.6 to 8.3)

PFS at 12 months, % (95% CI)

NA

NA

43.0

(34.6 to 51.0)

25.7

(18.6 to 33.4)

PFS at 24 months, % (95% CI)

NA

NA

23.4

(16.0 to 31.7)

12.6

(7.3 to 19.5)

Hazard ratio (95% CI)

NA

NA

0.62 (0.48 to 0.82)

Overall survival

Number of events, n (%)

NR

11 (15.9)

13 (16.0)

9 (15.0)

||||||

||||

Median OS, months (95% CI)

NR

Not reached

Not reached

Not reached

||||||

||||

OS at 12 months, % (95% CI)

100 (NR)

95.7 (NR)

96.2 (NR)

96.6 (NR)

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

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

OS at 24 months, % (95% CI)

NR

92.6 (NR)

91.0 (NR)

89.4 (NR)

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

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

Hazard ratio (95% CI)

NA

NA

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

Objective response rate

Number of responses, n (%)

3 (42.9)

54 (78.3)

52 (64.2)

43 (71.7)

99 (65.6)

83 (54.3)

   Complete response

2 (28.6)

18 (26.1)

21 (25.9)

19 (31.7)

37 (24.5)

37 (24.2)

   Partial response

1 (14.3)

36 (52.2)

31 (38.3)

24 (40.0)

62 (41.1)

46 (30.1)

   Stable disease

3 (42.9)

8 (11.6)

8 (9.9)

7 (11.7)

21 (13.9)

36 (23.5)

   Progressive disease

1 (14.3)

5 (7.2)

19 (23.5)

10 (16.7)

26 (17.2)

28 (18.3)

   Not evaluable

0 (0.0)

NR

NR

NR

1 (0.7)

1 (0.7)

   No assessment

0 (0.0)

2 (2.9)

2 (2.5)

0 (0.0)

4 (2.6)

5 (3.3)

Proportion with a response, % (95% CI); P value

42.9

(9.9 to 81.6)

78.3

(66.7 to 87.3)

P < 0.001a

64.2

(52.8 to 74.6)

P < 0.001a

71.7

(58.6 to 82.5)

P < 0.001a

65.6

(57.4 to 73.1)

54.2

(46.0 to 62.3)

Difference in response rate (95% CI)

NA

NA

11.3 (0.2 to 22.1)

P = 0.022534b

Proportion with complete response, % (95% CI)

28.6 (3.7 to 71.0)

26.1 (16.3 to 38.1)

25.9 (16.8 to 36.9)

31.7 (20.3 to 45.0)

24.5 (17.9 to 32.2)

24.2 (17.6 to 31.8)

Duration of response

Median duration, months (range)

Not reached

(0.0 to 6.1)

25.0

(0.0 to 36.1)

11.1

(0.0 to 35.9)

16.8

(0.0 to 39.1)

20.7

(0.0 to 33.2)

13.8

(0.0 to 33.9)

Patients with extended duration of response, n (%)

   ≥ 6 months

1 (50.0)

34 (81.1)

23 (68.9)

27 (72.7)

66 (79.9)

34 (59.6)

   ≥ 12 months

NR

27 (66.5)

14 (44.4)

19 (64.1)

48 (62.4)

23 (50.0)

   ≥ 18 months

NR

NR

NR

NR

31 (53.7)

13 (42.8)

   ≥ 24 months

NR

15 (50.4)

10 (34.2)

11 (49.8)

11 (47.4)

7 (42.8)

Time to response

Mean, months (SD)

2.5 (0.4)

3.4 (1.8)

3.2 (1.4)

4.2 (2.8)

3.7 (3.9)

2.9 (0.6)

Median, months (range)

2.6

(2.1 to 2.8)

2.7

(2.1 to 12.9)

2.8

(2.2 to 11.0)

2.8

(2.6 to 16.5)

2.8

(1.0 to 31.2)

2.8

(1.3 to 7.3)

Health-related quality of lifec

Change in LS mean EORTC QLQ-C30 Global Health Status between week 24 and baseline, mean (95% CI)

NR

11.8 (NR)d

13.9 (NR)e

6.6 (NR)f

7.29

(3.94 to 10.64)

–1.31

(–5.17 to 2.55)

Difference in LS mean change on EORTC QLQ-C30 Global Health Status, (95% CI)

NR

NR

8.60 (3.89 to 13.31)

LS mean change in EORTC QLQ-C30 Physical Functioning Scale between week 24 and baseline, mean (95% CI)

NR

NR

4.31

(1.15 to 7.47)

–1.93

(–5.44 to 1.58)

Difference in LS mean change on EORTC QLQ-C30 Physical Functioning Scale, (95% CI)

NR

NR

6.24 (1.87 to 10.62)

Change in LS mean EQ-5D-3L Utility Score between week 24 and baseline, mean (95% CI)

NR

NR

0.04

(0.00 to 0.08)

–0.05

(–0.09 to –0.01)

Difference in LS mean change on EQ-5D-3L Utility Score, (95% CI)

NR

NR

0.09 (0.04 to 0.14)

LS mean change in EQ-5D-3L visual analogue scale between week 24 and baseline, mean (95% CI)

NR

NR

8.53

(5.42 to 11.64)

2.41

(–1.05 to 5.87)

Difference in LS mean change on EQ-5D-3L visual analogue scale, (95% CI)

NR

NR

6.12 (1.91 to 10.34)

CI = confidence interval; EORTC QLQ-C30 = European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30; EQ-5D-3L = EuroQol 5-Dimensions 3-Levels questionnaire; LS = least squares; NA = not applicable; NR = not reported; OS = overall survival; PFS = progression-free survival; SD = standard deviation.

aStatistically significant.

bStatistically insignificant.

cOnly 146 and 150 pembrolizumab and brentuximab vedotin patients, respectively, in the KEYNOTE-204 study had complete EORTC data.

dOnly 69 individuals had complete data.

eOnly 79 individuals had complete data.

fOnly 58 individuals had complete data.

Source: Clinical Study Reports for the KEYNOTE-051, KEYNOTE-087, and KEYNOTE-204 studies.

Harms

The safety outcomes (harms) identified in the review protocol are reported below. See Table 24 for detailed harms data.

Adverse Events

In the KEYNOTE-051 study, 85.7% of patients experienced at least 1 AE. In the KEYNOTE-087 study, 98.6%, 98.8%, and 95.0% of patients experienced at least 1 AE in cohort 1, cohort 2, and cohort 3, respectively. In the KEYNOTE-204 study, 98.0% of patients in the pembrolizumab arm and 94.1% of those in the BV arm experienced an AE. The most common AEs were pyrexia, vomiting, headache, abdominal pain, anemia, cough, fatigue, diarrhea, and upper respiratory tract infections. In the KEYNOTE-204 study, pembrolizumab patients were more likely than BV patients to experience endocrine disorders (20.3% versus 3.9%); infections (66.2% versus 45.4%); musculoskeletal and connective tissue disorders (37.8% versus 31.6%); neoplasms (7.4% versus 1.3%), renal or urinary disorders (14.9% versus 4.6%); respiratory, thoracic, or mediastinal disorders (45.3% versus 26.3%); and skin and subcutaneous tissue disorders (43.9% versus 36.8%), but less likely to experience blood or lymphatic system disorders (18.2% versus 25.7%), gastrointestinal disorders (43.9% versus 52.0%), and nervous system disorders (26.4% versus 50.7%).

Serious Adverse Events

In the KEYNOTE-051 study, 28.6% of patients experienced at least 1 serious AE. In the KEYNOTE-087 study, 21.7%, 22.2%, and 25.0% of patients experienced a serious AE in cohort 1, cohort 2, and cohort 3, respectively. In the KEYNOTE-204 study, 29.7% of pembrolizumab and 21.1% of BV-treated patients experienced a serious AE. The most common serious AEs in the KEYNOTE-051 study were diaphragmatic hernia and pneumonia. The most common serious AEs in cohort 1 of the KEYNOTE-087 study were pneumonia and pericarditis. The most common serious AE in cohort 2 of the KEYNOTE-087 study was herpes zoster and the most common serious AEs in cohort 3 of the KEYNOTE-087 study were pyrexia and pneumonitis, There were no notable differences in frequency of serious AEs between the pembrolizumab and BV arms in the KEYNOTE-204 study. The most common serious AEs in the pembrolizumab arm of the KEYNOTE-204 study were infections or infestations; respiratory, thoracic, or mediastinal disorders; neoplasms; general disorders or administration site conditions; and hepatobiliary disorders. The most common serious AEs in the BV arm of the KEYNOTE-204 study were infections or infestations; respiratory, thoracic, or mediastinal disorders; nervous system disorders; gastrointestinal disorders; and general disorders or administration site conditions.

Treatment Discontinuation Due to AEs

No patients in KEYNOTE-051 discontinued treatment due to an AE while 11.6%, 6.2%, and 8.3% of patients in cohort 1, cohort 2, and cohort 3 of KEYNOTE-087, respectively, discontinued treatment due to an AE. In the KEYNOTE-204 study, 13.5% and 17.8% of patients receiving pembrolizumab and BV, respectively, discontinued treatment due to an AE.

Mortality

No patients in the KEYNOTE-051 study or cohort 1 of the KEYNOTE-087 study died. In the KEYNOTE-087 study, 2.5% and 1.7% of patients in cohort 2 and cohort 3, respectively, died. In the KEYNOTE-204 study, 2.0% and 1.3% of patients receiving pembrolizumab or BV, respectively, died.

Immune-Mediated AEs

In the KEYNOTE-051 study, 28.6% of patients experienced at least 1 immune-mediated AE. In cohort 1, 2, and 3 of the KEYNOTE-087 study, 31.9%, 32.1%, and 38.3% of patients, respectively, experienced at least 1 immune-mediated AE. In the KEYNOTE-204 study, more patients in the pembrolizumab arm (35.8%) than the BV arm (13.8%) experienced an immune-mediated AE.

No patients in the KEYNOTE-051 study experienced a serious immune-mediated AE. In the KEYNOTE-087 study, 4.3%, 2.5% and 5.0% of patients in cohort 1, cohort 2, and cohort 3, respectively, experienced a serious immune-mediated AE. In the KEYNOTE-204 study, more pembrolizumab- than BV-treated patients experienced a serious immune-mediated AE (8.8% versus 3.3%).

Table 24: Summary of Harms

Harms, n (%)

KEYNOTE-051

KEYNOTE-087

KEYNOTE-204

Pembrolizumab

N = 7

Cohort 1

N = 69

Cohort 2

N = 81

Cohort 3

N = 60)

Pembrolizumab

N = 148

Brentuximab vedotin

N = 152

Patients with at least 1 adverse event

6 (85.7)

68 (98.6)

80 (98.8)

57 (95.0)

145 (98.0)

143 (94.1)

Patients with at least 1 serious adverse event

2 (28.6)

15 (21.7)

18 (22.2)

15 (25.0)

44 (29.7)

32 (21.1)

Patients who discontinued treatment due to adverse event

0 (0.0)

8 (11.6)

5 (6.2)

5 (8.3)

20 (13.5)

27 (17.8)

Patients who died

0 (0.0)

0 (0.0)

2 (2.5)

1 (1.7)

3 (2.0)

2 (1.3)

Patients with at least 1 immune-mediated adverse event

2 (28.6)

22 (31.9)

26 (32.1)

23 (38.3)

53 (35.8)

21 (13.8)

Patients with at least 1 grade III to V immune-mediated adverse event

0 (0.0)

3 (4.3)

4 (4.9)

1 (1.7)

11 (7.4)

5 (3.3)

Patients with at least 1 serious immune-mediated adverse event

0 (0.0)

3 (4.3)

2 (2.5)

3 (5.0)

13 (8.8)

5 (3.3)

Note: Values are indicated as n (%).

Source: Clinical Study Reports for the KEYNOTE-051, 87, and 204 studies .

Table 25: Most Frequent Adverse Events in KEYNOTE-051

Adverse event, n (%)

Pembrolizumab

N = 7

Abdominal pain

4 (57.1)

Headache

4 (57.1)

Pyrexia

4 (57.1)

Vomiting

4 (57.1)

Cough

3 (42.9)

Diarrhea

3 (42.9)

Hyperglycemia

3 (42.9)

Arthralgia

2 (28.6)

Decreased appetite

2 (28.6)

Hemoglobin decreased

2 (28.6)

Hypernatremia

2 (28.6)

Musculoskeletal pain

2 (28.6)

Nasal congestion

2 (28.6)

Neutropenia

2 (28.6)

Pneumonia

2 (28.6)

White blood cells decreased

2 (28.6)

Alanine aminotransferase increased

1 (14.3)

Aspartate aminotransferase increased

1 (14.3)

Asthenia

1 (14.3)

Axillary pain

1 (14.3)

Back pain

1 (14.3)

Blood albumin decreased

1 (14.3)

Blood alkaline phosphatase increased

1 (14.3)

Blood calcium decreased

1 (14.3)

Blood magnesium decreased

1 (14.3)

Blood thyroid stimulating hormone increased

1 (14.3)

Blood uric acid increased

1 (14.3)

Bradycardia

1 (14.3)

Chest pain

1 (14.3)

Chills

1 (14.3)

Constipation

1 (14.3)

Dermal cyst

1 (14.3)

Diaphragmatic hernia

1 (14.3)

Fatigue

1 (14.3)

Gastritis

1 (14.3)

Hyperhidrosis

1 (14.3)

Hyperthyroidism

1 (14.3)

Hypoalbuminemia

1 (14.3)

Hypothyroidism

1 (14.3)

Leukopenia

1 (14.3)

Lymphopenia

1 (14.3)

Memory impairment

1 (14.3)

Menstruation irregular

1 (14.3)

Myalgia

1 (14.3)

Nasopharyngitis

1 (14.3)

Nausea

1 (14.3)

Neck pain

1 (14.3)

Neuralgia

1 (14.3)

Night sweats

1 (14.3)

Oropharyngeal pain

1 (14.3)

Pain in extremity

1 (14.3)

Papule

1 (14.3)

Pneumonitis

1 (14.3)

Procedural pain

1 (14.3)

Proteinuria

1 (14.3)

Scab

1 (14.3)

Sinus tachycardia

1 (14.3)

Skin abrasion

1 (14.3)

Sleep disorder

1 (14.3)

Tachycardia

1 (14.3)

Thrombocytopenia

1 (14.3)

Upper respiratory tract infection

1 (14.3)

Urinary tract infection

1 (14.3)

Urticaria

1 (14.3)

Vitiligo

1 (14.3)

Table 26: Most Frequent Adverse Events in KEYNOTE-087; Incidence of 10% or Greater in 1 or More Groups

Adverse event, n (%)

Cohort 1

N = 69

Cohort 2

N = 81

Cohort 3

N = 80

Pyrexia

27 (39.1)

19 (23.5)

17 (28.3)

Cough

19 (27.5)

22 (27.2)

14 (23.3)

Fatigue

15 (21.7)

17 (21.0)

16 (26.7)

Diarrhea

20 (29.0)

12 (14.8)

11 (18.3)

Upper respiratory infection

23 (33.3)

7 (8.6)

13 (21.7)

Nausea

16 (23.2)

11 (13.6)

11 (18.3)

Vomiting

16 (23.2)

9 (11.1)

13 (21.7)

Nasopharyngitis

12 (17.4)

16 (19.8)

7 (11.7)

Hypothyroidism

8 (11.6)

13 (16.0)

12 (20.0)

Rash

12 (17.4)

9 (11.1)

10 (16.7)

Pruritus

12 (17.4)

11 (13.6)

7 (11.7)

Headache

12 (17.4)

6 (7.4)

10 (16.7)

Arthralgia

10 (14.5)

10 (12.3)

7 (11.7)

Backpain

8 (11.6)

12 (14.8)

5 (8.3)

Dyspnea

8 (11.6)

10 (12.3)

7 (11.7)

Constipation

9 (13.0)

11 (13.6)

3 (5.0)

Oropharyngeal pain

7 (10.1)

7 (8.6)

9 (15.0)

Nasal congestion

6 (8.7)

8 (9.9)

8 (13.3)

Anemia

8 (11.6)

8 (9.9)

5 (8.3)

Sinusitis

7 (10.1)

9 (11.1)

5 (8.3)

Insomnia

7 (10.1)

6 (7.4)

7 (11.7)

Bronchitis

6 (8.7)

5 (6.2)

7 (11.7)

Asthenia

6 (8.7)

9 (11.1)

2 (3.3)

Rhinorrhea

9 (13.0)

4 (4.9)

3 (5.0)

Productive cough

10 (14.5)

1 (1.2)

4 (6.7)

Muscle spasms

8 (11.6)

1 (1.2)

5 (8.3)

Alanine aminotransferase increased

7 (10.1)

3 (3.7)

0 (0.0)

Rhinitis

8 (11.6)

0 (0.0)

2 (3.3)

Influenza like illness

7 (10.1)

2 (2.5)

0 (0.0)

Table 27: Most Frequent Adverse Events in KEYNOTE-204; Incidence of 5% or Greater in 1 or More Groups

Adverse event, n (%)

Pembrolizumab

N = 148

Brentuximab vedotin

N = 152

Blood and lymphatic system disorders

27 (18.2)

39 (25.7)

   Anemia

9 (6.1)

13 (8.6)

   Neutropenia

10 (6.8)

20 (13.2)

   Thrombocytopenia

9 (6.1)

8 (5.3)

Cardiac disorders

8 (5.4)

3 (2.0)

Endocrine disorders

30 (20.3)

6 (3.9)

   Hyperthyroidism

8 (5.4)

1 (0.7)

   Hypothyroidism

28 (18.9)

4 (2.6)

Eye disorders

9 (6.1)

5 (3.3)

Gastrointestinal disorders

65 (43.9)

79 (52.0)

   Abdominal pain

10 (6.8)

15 (9.9)

   Constipation

11 (7.4)

19 (12.5)

   Diarrhea

29 (19.6)

25 (16.4)

   Dyspepsia

7 (4.7)

9 (5.9)

   Nausea

21 (14.2)

37 (24.3)

   Vomiting

20 (13.5)

30 (19.7)

General disorders

64 (43.2)

59 (38.8)

   Asthenia

8 (5.4)

7 (4.6)

   Fatigue

23 (15.5)

28 (18.4)

   Pyrexia

29 (19.6)

20 (13.2)

Hepatobiliary disorders

8 (5.4)

3 (2.0)

Infections

98 (66.2)

69 (45.4)

   Nasopharyngitis

17 (11.5)

8 (5.3)

   Pneumonia

12 (8.1)

9 (5.9)

   Rhinitis

8 (5.4)

5 (3.3)

   Sinusitis

8 (5.4)

3 (2.0)

   Upper respiratory tract infection

28 (18.9)

22 (14.5)

   Urinary tract infection

16 (10.8)

4 (2.6)

Injury, poisoning, and procedural complications

17 (11.5)

19 (12.5)

   Infusion-related reactions

5 (3.4)

12 (7.9)

Investigations

46 (31.1)

39 (25.7)

   Alanine aminotransferase increased

13 (8.8)

15 (9.9)

   Aspartate aminotransferase increased

12 (8.1)

11 (7.2)

   Neutrophil count decreased

3 (2.0)

10 (6.6)

   Weight decrease

5 (3.4)

11 (7.2)

   Weight increase

10 (6.8)

2 (1.3)

Metabolism and nutritional disorders

30 (20.3)

29 (19.1)

   Decreased appetite

9 (6.1)

14 (9.2)

Musculoskeletal and connective tissue disorders

56 (37.8)

48 (31.6)

   Arthralgia

13 (8.8)

11 (7.2)

   Back pain

19 (12.8)

18 (11.8)

   Myalgia

7 (4.7)

10 (6.6)

   Pain in extremity

13 (8.8)

7 (4.6)

Neoplasms

11 (7.4)

2 (1.3)

   Anogenital warts

0 (0.0)

1 (0.7)

   B-cell lymphoma

1 (0.7)

0 (0.0)

   Fibroma

1 (0.7)

0 (0.0)

   Pancreatic neuroendocrine tumour

1 (0.7)

0 (0.0)

   Skin papilloma

1 (0.7)

0 (0.0)

   Squamous cell carcinoma

1 (0.7)

0 (0.0)

   Squamous cell carcinoma of the cervix

1 (0.7)

0 (0.0)

   Tumour flare

2 (1.4)

0 (0.0)

   Tumour pain

3 (2.0)

0 (0.0)

   Tumour ulceration

0 (0.0)

1 (0.7)

Nervous system disorders

39 (26.4)

77 (50.7)

   Headache

15 (10.1)

15 (9.9)

   Neuropathy, peripheral

6 (4.1)

28 (18.4)

   Paresthesia

7 (4.7)

10 (6.6)

   Peripheral sensory neuropathy

4 (2.7)

21 (13.8)

Psychiatric disorders

21 (14.2)

27 (17.8)

   Anxiety

7 (4.7)

12 (7.9)

Renal and urinary disorders

22 (14.9)

7 (4.6)

Reproductive system and breast disorders

11 (7.4)

4 (2.6)

Respiratory, thoracic, and mediastinal disorders

67 (45.3)

40 (26.3)

   Cough

25 (16.9)

20 (13.2)

   Dyspnea

11 (7.4)

9 (5.9)

   Oropharyngeal pain

12 (8.1)

5 (3.3)

   Pneumonitis

13 (8.8)

3 (2.0)

Skin and subcutaneous tissue disorders

65 (43.9)

56 (36.8)

   Pruritus

26 (17.6)

18 (11.8)

   Rash

13 (8.8)

13 (8.6)

Vascular disorders

14 (9.5)

10 (6.6)

Table 28: Most Frequent Serious Adverse Events in KEYNOTE-051

Serious adverse event, n (%)

Pembrolizumab

N = 7

Diaphragmatic hernia

1 (14.3)

Pneumonia

1 (14.3)

Table 29: Most Frequent Serious Adverse Events in KEYNOTE-087; Incidence of 1% or Greater

Serious adverse event, n (%)

Cohort 1

N = 69

Cohort 2

N = 81

Cohort 3

N = 80

Pneumonia

4 (5.8)

1 (1.2)

1 (1.7)

Pneumonitis

1 (1.4)

1 (1.2)

2 (3.3)

Pyrexia

0 (0.0)

1 (1.2)

3 (5.0)

Acute graft vs. host disease

1 (1.4)

1 (1.2)

1 (1.7)

Bronchitis

0 (0.0)

1 (1.2)

1 (1.7)

Herpes zoster

0 (0.0)

2 (2.5)

0 (0.0)

Pericarditis

2 (2.9)

0 (0.0)

0 (0.0)

Acute kidney injury

1 (1.4)

0 (0.0)

0 (0.0)

Acute sinusitis

0 (0.0)

1 (1.2)

0 (0.0)

Anemia

1 (1.4)

0 (0.0)

0 (0.0)

Aortic stenosis

0 (0.0)

0 (0.0)

1 (1.7)

Autoimmune hepatitis

1 (1.4)

0 (0.0)

0 (0.0)

Basal cell carcinoma

0 (0.0)

1 (1.2)

0 (0.0)

Blood creatine phosphokinase increase

0 (0.0)

1 (1.2)

0 (0.0)

Bowen disease

0 (0.0)

1 (1.2)

0 (0.0)

Bronchopulmonary aspergillosis

0 (0.0)

1 (1.2)

0 (0.0)

Chronic inflammatory demyelinating polyradiculoneuropathy

0 (0.0)

1 (1.2)

0 (0.0)

Clostridium difficile colitis

0 (0.0)

1 (1.2)

0 (0.0)

Cystitis

0 (0.0)

0 (0.0)

1 (1.7)

Cytokine release syndrome

0 (0.0)

1 (1.2)

0 (0.0)

Device-related infection

0 (0.0)

1 (1.2)

0 (0.0)

Diarrhea

1 (1.4)

0 (0.0)

0 (0.0)

Diffuse large B-cell lymphoma

0 (0.0)

1 (1.2)

0 (0.0)

Epilepsy

0 (0.0)

0 (0.0)

1 (1.7)

Escherichia bacteremia

0 (0.0)

1 (1.2)

0 (0.0)

Gastroenteritis

0 (0.0)

0 (0.0)

1 (1.7)

Gastroenteritis salmonella

0 (0.0)

1 (1.2)

0 (0.0)

Herpes simplex

1 (1.4)

0 (0.0)

0 (0.0)

Hip fracture

1 (1.4)

0 (0.0)

0 (0.0)

Hypersensitivity

0 (0.0)

0 (0.0)

1 (1.7)

Hyperthermia

0 (0.0)

1 (1.2)

0 (0.0)

Influenza

1 (1.4)

0 (0.0)

0 (0.0)

Infusion-related reaction

0 (0.0)

1 (1.2)

0 (0.0)

Lower respiratory tract infection

0 (0.0)

0 (0.0)

1 (1.7)

Lung infection

0 (0.0)

1 (1.2)

0 (0.0)

Myelitis

1 (1.4)

0 (0.0)

0 (0.0)

Myelodysplastic syndrome

1 (1.4)

0 (0.0)

0 (0.0)

Myocardial infarction

0 (0.0)

0 (0.0)

1 (1.7)

Myocarditis

1 (1.4)

0 (0.0)

0 (0.0)

Necrotizing myositis

1 (1.4)

0 (0.0)

0 (0.0)

Osteonecrosis

1 (1.4)

0 (0.0)

0 (0.0)

Pneumothorax

0 (0.0)

0 (0.0)

1 (1.7)

Polyneuropathy

1 (1.4)

0 (0.0)

0 (0.0)

Post-procedural infection

0 (0.0)

1 (1.2)

0 (0.0)

Pulmonary embolism

0 (0.0)

1 (1.2)

0 (0.0)

Respiratory syncytial virus infection

0 (0.0)

0 (0.0)

1 (1.7)

Respiratory tract infection

1 (1.4)

0 (0.0)

0 (0.0)

Schizophrenia

0 (0.0)

0 (0.0)

1 (1.7)

Septic shock

0 (0.0)

1 (1.2)

0 (0.0)

Small cell lung cancer

1 (1.4)

0 (0.0)

0 (0.0)

Squamous cell carcinoma

0 (0.0)

1 (1.2)

0 (0.0)

Stress cardiomyopathy

0 (0.0)

1 (1.2)

0 (0.0)

Upper respiratory tract infection

0 (0.0)

0 (0.0)

1 (1.7)

Urosepsis

1 (1.4)

0 (0.0)

0 (0.0)

Varicella zoster virus infection

1 (1.4)

0 (0.0)

0 (0.0)

Table 30: Most Frequent Serious Adverse Events in KEYNOTE-204; Incidence of 1% or Greater

Serious adverse event, n (%)

Pembrolizumab

N = 148

Brentuximab vedotin

N = 152

Blood and lymphatic system disorders

4 (2.7)

1 (0.7)

   Febrile neutropenia

2 (1.4)

0 (0.0)

Cardiac disorders

3 (2.0)

1 (0.7)

   Myocarditis

2 (1.4)

0 (0.0)

Gastrointestinal disorders

2 (1.4)

4 (2.6)

General disorders and administration site conditions

5 (3.4)

3 (2.0)

   Pyrexia

4 (2.7)

1 (0.7)

Hepatobiliary disorders

4 (2.7)

0 (0.0)

Infections and infestations

18 (12.2)

12 (7.9)

   Pneumonia

8 (5.4)

5 (3.3)

Injury, poisoning, and procedural complications

1 (0.7)

3 (2.0)

   Infusion-related reaction

0 (0.0)

3 (2.0)

Neoplasms benign, malignant, and unspecified

6 (4.1)

0 (0.0)

Nervous system disorders

1 (0.7)

5 (3.3)

   Neuropathy peripheral

0 (0.0)

2 (1.3)

Renal and urinary disorders

2 (1.4)

1 (0.7)

   Acute kidney injury

2 (1.4)

0 (0.0)

Respiratory, thoracic, and mediastinal disorders

11 (7.4)

6 (3.9)

   Interstitial lung disease

2 (1.4)

1 (0.7)

   Pneumonitis

8 (5.4)

1 (0.7)

   Pulmonary embolism

0 (0.0)

2 (1.3)

Skin and subcutaneous tissue disorders

1 (0.7)

2 (1.3)

Vascular disorders

2 (1.4)

1 (0.7)

Table 31: Most Frequent Immune-Mediated Adverse Events in KEYNOTE-051

Immune-mediated adverse event, n (%)

Pembrolizumab

N = 7

Endocrine disorders

2 (28.6)

   Hyperthyroidism

1 (14.3)

   Hypothyroidism

1 (14.3)

Respiratory, thoracic, and mediastinal disorders

1 (14.3)

   Pneumonitis

1 (14.3)

Table 32: Most Frequent Immune-Mediated Adverse Events in KEYNOTE-087

Immune-mediated adverse event, n (%)

Cohort 1

N = 69

Cohort 2

N = 81

Cohort 3

N = 80

Colitis

2 (2.9)

1 (1.2)

0 (0.0)

   Colitis

1 (1.4)

1 (1.2)

0 (0.0)

   Enterocolitis

1 (1.4)

0 (0.0)

0 (0.0)

Encephalitis

0 (0.0)

0 (0.0)

1 (1.7)

Hepatitis, autoimmune

1 (1.4)

0 (0.0)

0 (0.0)

Hyperthyroidism

1 (1.4)

4 (4.9)

3 (5.0)

Hypothyroidism

8 (11.6)

13 (16.0)

12 (20.0)

Infusion reactions

8 (11.6)

6 (7.4)

8 (13.3)

   Cytokine release syndrome

0 (0.0)

4 (4.9)

2 (3.3)

   Drug hypersensitivity

0 (0.0)

0 (0.0)

1 (1.7)

   Hypersensitivity

3 (4.3)

0 (0.0)

3 (5.0)

   Infusion-related reaction

6 (8.7)

3 (3.7)

2 (3.3)

Myocarditis

1 (1.4)

0 (0.0)

0 (0.0)

Myositis

2 (2.9)

0 (0.0)

0 (0.0)

   Myositis

1 (1.4)

0 (0.0)

0 (0.0)

   Necrotizing myositis

1 (1.4)

0 (0.0)

0 (0.0)

Pneumonitis

3 (4.3)

4 (4.9)

4 (6.7)

   Organizing pneumonia

0 (0.0)

1 (1.2)

0 (0.0)

   Pneumonitis

3 (4.3)

3 (3.7)

4 (6.7)

Sarcoidosis

0 (0.0)

1 (1.2)

0 (0.0)

Skin

1 (1.4)

1 (1.2)

0 (0.0)

   Lichen planus

0 (0.0)

1 (1.2)

0 (0.0)

   Pruritus

1 (1.4)

0 (0.0)

0 (0.0)

Thyroiditis, autoimmune

0 (0.0)

0 (0.0)

1 (1.7)

Uveitis

3 (4.3)

0 (0.0)

0 (0.0)

   Chorioretinitis

1 (1.4)

0 (0.0)

0 (0.0)

   Iridocyclitis

1 (1.4)

0 (0.0)

0 (0.0)

   Iritis

1 (1.4)

0 (0.0)

0 (0.0)

Table 33: Most Frequent Immune-Mediated Adverse Events in KEYNOTE-204

Immune-mediated adverse event, n (%)

Pembrolizumab

N = 148

Brentuximab vedotin

N = 152

Adrenal Insufficiency

1 (0.7)

0 (0.0)

Colitis

1 (0.7)

1 (0.7)

Encephalitis

1 (0.7)

0 (0.0)

Hepatitis

1 (0.7)

0 (0.0)

Hyperthyroidism

8 (5.4)

1 (0.7)

Hypothyroidism

28 (18.9)

4 (2.6)

Infusion reactions

8 (5.4)

12 (7.9)

   Drug hypersensitivity

1 (0.7)

0 (0.0)

   Hypersensitivity

2 (1.4)

0 (0.0)

   Infusion-related reaction

5 (3.4)

12 (7.9)

Myocarditis

2 (1.4)

0 (0.0)

Myositis

1 (0.7)

0 (0.0)

Nephritis

1 (0.7)

1 (0.7)

   Nephritis

1 (0.7)

0 (0.0)

   Tubulointerstitial nephritis

0 (0.0)

1 (0.7)

Pancreatitis

2 (1.4)

0 (0.0)

Pneumonitis

16 (10.8)

4 (2.6)

   Interstitial lung disease

3 (2.0)

1 (0.7)

   Pneumonitis

13 (8.8)

3 (2.0)

Severe skin reactions

3 (2.0)

3 (2.0)

   Dermatitis exfoliative

0 (0.0)

1 (0.7)

   Dermatitis exfoliative, generalized

0 (0.0)

1 (0.7)

   Pruritus, genital

1 (0.7)

0 (0.0)

   Toxic skin eruption

2 (1.4)

1 (0.7)

Thyroiditis

2 (1.4)

0 (0.0)

Uveitis

2 (1.4)

0 (0.0)

Critical Appraisal

Internal Validity of KEYNOTE-051

The KEYNOTE-051 study originally did not aim to specifically recruit patients with relapsed or refractory cHL. A study protocol amendment was made to identify patients who clearly had relapsed or refractory cHL which then identified 7 patients. Only 3 response events were observed, based on these 7 patients. Due to the low event rate and sample size, it is uncertain if these results alone are representative of the potential benefits and harms of pembrolizumab. The KEYNOTE-051 study was also an open-label trial without a comparator, thus any incremental benefit over standard of care is unknown and fails to mitigate the impact of confounding variables, but the trial assessed response to therapy using an independent and blinded assessor which reduces any bias introduced by the open-label design on outcomes such as PFS, ORR, complete response rate, DOR, and time to response.

External Validity of KEYNOTE-051

The KEYNOTE-051 study did not explicitly recruit patients who failed ASCT or were ineligible for ASCT and salvage chemotherapy. Thus, generalizability of the KEYNOTE-051 study to the requested patient population is uncertain. These results may also only be applicable to those with a Lansky Play-Performance Scale score of 50 or greater for children 16 and younger or a Karnofsky score of 50 or greater in children aged 16 years and older.

Internal Validity of KEYNOTE-087

The KEYNOTE-087 study is a single-arm, open-label trial and thus provides limited insight on any additional benefit over the current standard of care. The open-label nature of this trial may have biased the HRQoL assessment, but the trial did assess response to therapy using an independent and blinded assessor which reduces the bias introduced by the open-label design on outcomes such as PFS, ORR, complete response rate, DOR, and time to response.

External Validity of KEYNOTE-087

The results from the KEYNOTE-087 study are generalizable to most adult patients who have failed an ASCT or are ineligible for ASCT and salvage chemotherapy. The KEYNOTE-087 study excluded those with an ECOG performance score of 2 or greater; hence, its results may not be generalizable to this population. Notably, cohort 1 and 2 included patients who had received and failed on BV in addition to having a history of disease progression after ASCT (cohort 1) or being ineligible for ASCT (cohort 2), and thus do not completely align with the population of interest in this review.

Internal Validity of KEYNOTE-204

The KEYNOTE-204 study was an open-label trial which randomized patients centrally using an interactive voice response system and integrated web response system. Randomization was stratified based on prior ASCT (yes or no) and cHL status after first-line therapy (primary refractory, relapsed disease < 12 months after completion of first-line therapy, or relapse 12 months or longer after completing first-line therapy). Randomization helps to ensure prognostic balance at the start of the study and baseline characteristics were generally balanced between arms suggesting randomization was successful. There were slightly higher rates of bulky disease, baseline B symptoms, and baseline bone marrow involvement in the pembrolizumab arm; however, the clinical experts do not believe this imbalance detracts from the results favouring pembrolizumab as the patients in the pembrolizumab arm had more adverse baseline characteristics than those in the BV arm.

The KEYNOTE-204 study allowed patients to receive subsequent anticancer therapies once a trial medication (pembrolizumab or BV) was discontinued but did not allow concurrent use of other anticancer treatments with trial medications. Almost all patients randomized to BV (97.4%) received a subsequent anticancer therapy while 70.2% of pembrolizumab-treated patients did so. Those randomized to BV were more likely to cross over and subsequently receive pembrolizumab (17.8% versus 1.4% of patients who were originally randomized to pembrolizumab are retreated with pembrolizumab). Those originally randomized to BV were also more likely to receive nivolumab (19.7%) relative to those randomized to pembrolizumab (3.4%). Finally, 25.0% of patients originally randomized to pembrolizumab received BV while 4.6% of patients originally randomized to BV were retreated with BV (Table 19 and Appendix 2). Because these therapies were not used concurrently with a trial medication and were only used following a PFS event and after the trial medication was discontinued, it should not substantially impact the PFS analysis. However, subsequent utilization of anticancer therapies was considered a censoring event in the primary PFS analysis which may be a questionable assumption. In a pre-specified sensitivity analysis, initiation of subsequent anticancer therapies was considered a progression event and the resulting hazard ratio and 95% CI did not change substantially from the main analysis, suggesting the impact of subsequent anticancer therapy utilization on PFS might be limited. Subsequent utilization of anticancer therapies can impact the OS analysis; however, the use of subsequent anticancer medications was not evaluated by rank-preserving structural failure time or 2-stage analysis as suggested in the study protocol; thus, it is unclear what effect, if any, utilization of subsequent anticancer therapies would have on the OS results. |||||||||||||||||||||| |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||||||| |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| While the sponsor confirmed the proportional hazards assumption likely was not violated in the PFS analysis, it is unclear if the proportional hazards assumption was met in the model of OS as the assumption was not tested.

Treatment discontinuation rates were higher in the BV (96.1%) arm than in the pembrolizumab arm (74.3%) mostly due to higher discontinuations due to AEs and progressive disease. Discontinuation from the trial was also higher in the BV group (28.1% versus 13.2%) due to higher rates of death and withdrawal. These discontinuations may not be cause for methodological concern as they may reflect the superiority of pembrolizumab over BV. Alternatively, a propensity to discontinue therapy or the trial early could be influenced by the open-label nature of the study and a patient’s view of the medication they were randomized to. Similarly, the open-label nature of the study may have impacted subjective outcomes measures such as HRQoL. HRQoL was measured at baseline and week 24 but is unclear if at this point patients had already discontinued the trial medication and began utilization of a subsequent anticancer medication. If this is the case, their HRQoL scores may have been influenced by the subsequent anticancer medication and not the trial medication alone.

External Validity of KEYNOTE-204

The KEYNOTE-204 study results are generalizable to patients with cHL who failed ASCT or are ineligible for ASCT and multi-agent salvage chemotherapy. The KEYNOTE-204 study did not recruit individuals with an ECOG score of 2 or greater; therefore, results may not be applicable to this group. This study only compared pembrolizumab to BV, but not to any other comparators of interest listed in the study protocol. Notably, CADTH reviewed the use of BV in adults with HL after failure of at least 2 multi-agent chemotherapy regimens who are not candidates for ASCT and did not recommend reimbursement.35 However, the clinical experts consulted in the current review confirmed that in jurisdictions where it is funded, BV is still standard of care due to the lack of superior alternatives. This is in part supported by more recent evidence suggesting the efficacy of BV as third-line therapy in patients who have not received a stem cell transplant.36 Of note, BV is not universally funded across Canada at this time. The KEYNOTE-204 study did not conduct a subgroup analysis to analyze any differential impact of pembrolizumab on patients who failed ASCT and those who were ineligible for ASCT and salvage chemotherapy, thus the review team was unable to determine if there was a differential effect in each group. Approximately 11% of patients in each arm discontinued their study medication and subsequently received a stem cell transplant who were then censored in the PFS analysis at the time of transplant.

Table 34: Assessment of Generalizability of Evidence for Pembrolizumab

Domain

Factor

Evidence

CADTH’s assessment of generalizability

Population

Failed ASCT or ineligible for ASCT and salvage chemotherapy

KEYNOTE-204 and KEYNOTE-087

Due to a small event rate and sample size, the generalizability of KEYNOTE-051’s results are unclear. KEYNOTE-087 and KEYNOTE-204 are likely generalizable to the referenced population as the recruited population closely fits the sponsor’s requested indication except for some small constraints such as ECOG performance score.

Intervention

Pembrolizumab 200 mg every 3 weeks in adults and 2 mg/kg every 3 weeks in pediatrics

KEYNOTE-051, KEYNOTE-087, and KEYNOTE-204

These dosing regimens are generalizable to the requested dosing regimens.

Comparator

Brentuximab vedotin

KEYNOTE-204

The body of evidence may limit the generalizability to patients who would have otherwise been treated by brentuximab vedotin as pembrolizumab has not been compared with other alternatives.

Outcomes

PFS

OS

ORR and CR

DOR

HRQoL

KEYNOTE-051, KEYNOTE-087, and KEYNOTE-204

All listed outcomes are commonly studied and relevant to patients.

ASCT = autologous stem cell transplant; CR = complete response; DOR = duration of response; ECOG = Eastern Cooperative Oncology Group; HRQoL = health-related quality of life; ORR = objective response rate; OS = overall survival; PFS = progression-free survival.

Discussion

Summary of Available Evidence

One study, the KEYNOTE-051 study, assessed the efficacy and safety of 2 mg/kg of pembrolizumab administered every 3 weeks in 7 pediatric patients with relapsing or refractory cHL while 2 studies, the KEYNOTE-087 (N = 210) and KEYNOTE-204 (N = 304) studies, did so in the adults at a dose of 200 mg every 3 weeks. The KEYNOTE-087 study was a single-arm trial which divided patients in to 3 cohorts as shown below. Notably, cohorts 1 and 2 are similar but not identical to the population of interest as they have received and then failed or relapsed on BV. Patients in cohort 3 are similar to the population of interest as they had failed or relapsed after ASCT but were BV naive.

Finally, the KEYNOTE-204 study was an open-label, randomized trial comparing pembrolizumab (N = 151) with BV (N = 153) in patients with relapsed or refractory cHL who failed ASCT or were ineligible for ASCT but had trialled at least 2 multi-agent chemotherapy regimens. Although the KEYNOTE-204 study was an actively controlled trial, it is uncertain if the results observed could be extrapolated to other comparators.

Interpretation of Results

Efficacy

The KEYNOTE-051 study only recruited 7 pediatric patients; a sample size which is insufficient to be truly representative of pembrolizumab’s benefits and harms. The KEYNOTE-051 study was also an open-label, single-arm trial which limits the ability to estimate the incremental benefit of pembrolizumab over standard of care or mitigate the risk of confounding variables. Further, these patients had relapsed or refractory cHL but the study did not require patients to have a history of failure after ASCT, nor did it require patients to be ineligible for ASCT and multi-agent salvage therapy. Therefore, the KEYNOTE-051 study’s eligibility criteria do not completely align with the population of interest for this review. Due to the methodological limitations of the KEYNOTE-051 study, the conclusions made by the review team are mainly derived from the results reported in the KEYNOTE-087 and KEYNOTE-204 studies which only recruited adult patients. Among the cohorts of the KEYNOTE-087 study, cohort 3 most closely resembles the eligibility criteria for this review as these patients have failed ASCT and are BV naive while those in cohort 1 and 2 had trialled and failed BV.

Studies identified in this review do not provide a sufficient evidence base for a stand-alone reimbursement recommendation for the pediatric population. However, the clinical experts consulted by the review team confirmed that cHL is a disease which does not conform to the traditional and arguably artificial delineations of disease by age. Instead, cHL is often viewed as a disease with a similar biology and treatment approach, regardless of age. As such, while the median age of patients from the KEYNOTE-087 and KEYNOTE-204 studies ranges from 32.0 years to 40.0 years, the clinical experts believed these results were applicable to patients with cHL younger than 18 years of age. The clinical experts agreed that it might be biologically plausible to extrapolate the results of the KEYNOTE-087 and KEYNOTE-204 studies to pediatric patients and assume pembrolizumab may also benefit pediatric patients; however, evidence from studies with rigorous methodological quality are needed to confirm pembrolizumab’s benefits in the pediatric population. Further, it is uncertain if the pembrolizumab dosing regimen used in the KEYNOTE-087 and KEYNOTE-204 studies could be extrapolated to pediatric patients in whom the pharmacokinetic profile of pembrolizumab may differ. The KEYNOTE-051 study recruited 155 additional patients with other types of cancer, and in addition to past literature, pembrolizumab’s safety at this dose may be easier to establish but KEYNOTE-051 was a phase I/II study and thus it is unclear if 2 mg/kg is the most efficacious dose in pediatric patients. Such concerns would have been mitigated if the quality of evidence derived from the KEYNOTE-051 study was sufficiently robust to confirm that the effect of pembrolizumab administered at 2 mg/kg in pediatric patients was similar to the effect of pembrolizumab administered at 200 mg in adult patients. In the absence of this evidence, only clinical judgment and expertise can provide guidance on this issue.

Based on the KEYNOTE-087 and KEYNOTE-204 studies, pembrolizumab achieved clinically and statistically significant improvements over BV on PFS and clinically significant improvements on || ORR, DOR, and HRQoL. Patients treated with BV in the KEYNOTE-204 study had a median PFS of 8.3 months (95% CI, 5.7 to 8.8) while those treated with pembrolizumab had a median PFS of 13.2 months (95% CI, 10.9 to 19.4). The median PFS in cohort 3 of the KEYNOTE-087 study was the highest observed in all studies (19.4 months; 95% CI, 8.4 to 22.1) which supports the robust PFS evidence from the KEYNOTE-204 study. “Longer survival” was the most highly desired treatment outcome identified by patients. |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||  |  |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| PFS is a commonly used proxy indicator for OS in many oncology trials but CADTH staff were unable to identify any literature that quantified PFS power to predict OS in this patient population; thus, this report’s ability to translate pembrolizumab’s impact on PFS to OS will rely on clinical expertise.

ORR data were also encouraging as 11.3% (95% CI, 0.2 to 22.1) more patients in the pembrolizumab arm (65.6%; 95% CI, 57.4 to 73.1) of the KEYNOTE-204 study achieved a response relative to the BV arm (54.2%; 95% CI, 46.0 to 62.3). The proportion of patients in cohort 3 of the KEYNOTE-087 study achieving a response was even higher (71.7%; 95% CI, 58.6 to 82.5). However, the proportion of patients achieving a complete response was similar in the KEYNOTE-204 study regardless of treatment. CADTH’s clinical experts noted that while complete response rates are important, ORR is a more clinically relevant metric and patients may still have a relatively strong quality of life with a partial response or with disease stabilization. Patients also indicated that “longer remission” was a crucial treatment outcome. Relative to BV-treated patients, the DOR was higher in pembrolizumab-treated patients from the KEYNOTE-204 study and cohort 3 of the KEYNOTE-087 study.

Between baseline and week 24 in the KEYNOTE-204 study, HRQoL as measured by EORTC QLQ-C30 global health status improved by 8.60 (95% CI, 3.89 to 13.31) more points in the pembrolizumab arm relative to the BV arm. This is above the minimal important difference of 5 for cancer patients.45 Compared to the pembrolizumab arm of the KEYNOTE-204 study, EORTC QLQ-C30 global health status scores increased by a similar magnitude in cohort 3 of the KEYNOTE-087 study. Between baseline and week 24, HRQoL in pembrolizumab patients in the KEYNOTE-204 study improved more than BV patients when measured by the EQ-5D-3L utility scores (0.09; 95% CI, 0.04 to 0.14) and EQ-5D-3L visual analogue scores (6.12; 95% CI, 1.91 to 10.34) which exceeded the respective minimal important differences for patients with cancer of 0.05 and 6.47 While encouraging, HRQoL is a subjective measure which could have been influenced by the open-label nature of the trial and should be interpreted with caution. Finally, while minimal important differences were identified for patients with cancer, the minimal important differences were not specific to patients with cHL.

Harms

While BV patients in the KEYNOTE-204 study were least likely to experience an AE, serious AE, or immune-mediated AE, they were most likely to discontinue therapy due to an AE. There were no clear AEs that disproportionately affected the BV arm to an extent that would explain this phenomenon; hence, this observation cannot be clarified based on this data alone. One hypothesis is that BV-treated patients expected or observed fewer benefits and thus were less likely to tolerate the associated AEs and more likely to discontinue therapy. In this example, discontinuation is influenced by the fact that these patients were not blinded to their therapy and knew switching to pembrolizumab or another medication was an option. If they viewed BV as an inferior option, tolerance to AEs is decreased and propensity to discontinue BV in favour of another medication is increased. Conversely, pembrolizumab patients may be willing to tolerate more AEs and/or more serious AEs before discontinuing therapy if they felt pembrolizumab’s benefits, and hence remaining on pembrolizumab, was superior to other alternatives. Based on the feedback received from patients, 9 patients had previously been treated with pembrolizumab and generally spoke positively about pembrolizumab’s safety profile. Anecdotally, 7 of the 9 did not experience any negative impact on work or school and all reported a good to excellent experience with pembrolizumab, which is critical as “better quality of life,” and was the third most important treatment outcome to patients. The fourth most important outcome was fewer side effects and although BV generally had lower rates of AEs, 1 patient who trialled pembrolizumab and eventually discontinued it due to toxicity still stated the “PFS was worth the side effects.” Thus, from a patient’s perspective, pembrolizumab’s benefits may outweigh the harms while BV’s harms outweighed the benefits despite BV actually having fewer AEs.

Conclusions

The body of evidence included in this review suggests that, when compared to BV, pembrolizumab provides statistically and clinically significant improvement in PFS, as well as clinically significant improvements in || ORR, DOR, and HRQoL. |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||||||||||||||| Patients who received BV were generally less likely to experience AEs, serious AEs, or immune-mediated AEs, but more likely to discontinue therapy due to an AE. A definitive explanation of this phenomenon cannot be derived from this evidence alone. However, 1 explanation could be that BV-treated patients expected or observed worse health outcomes and thus were less willing to tolerate AEs, even if the rates were lower than in the pembrolizumab arm. Discontinuation would be a viable alternative for these patients as receiving another anticancer medication, including pembrolizumab, was an option. Conversely, pembrolizumab patients may have been willing to tolerate more AE as the expected benefits were commensurately higher. The body of evidence primarily evaluated pembrolizumab administered 200 mg every 3 weeks in adults but due to the nature of the disease, CADTH’s clinical experts believe that the benefits observed in adults would also be applicable to pediatric patients. However, because of insufficient evidence on the use of pembrolizumab in pediatric patients, it is uncertain what dose should be used to ascertain the benefits observed in adults. No other comparators to pembrolizumab aside from BV were evaluated in the included studies; thus, the comparative effect of pembrolizumab to other relevant treatments in the population under review, beyond BV, remains uncertain. Also, the KEYNOTE-087 and KEYNOTE-204 studies only recruited patients with an ECOG score of 0 or 1 but the CADTH clinical experts did not recommend limiting the use of pembrolizumab only to patients with low ECOG scores. In totality, the evidence suggests that pediatric and adult patients with relapsed or refractory cHL who failed ASCT or are ineligible for multi-agent salvage chemotherapy and ASCT are more likely to benefit from pembrolizumab than from BV; however, the dose required to ascertain these benefits in pediatrics is uncertain.

References

1.Canadian Cancer Society. Hodgkin lymphoma statistics. 2021; https://www.cancer.ca/en/cancer-information/cancer-type/hodgkin-lymphoma/statistics/?region=on. Accessed 2021 May 17.

2.Shenoy P, Maggioncalda A, Malik N, Flowers CR. Incidence patterns and outcomes for Hodgkin lymphoma patients in the United States. Adv Hematol. 2011;2011:725219. PubMed

3.Piris MA, Medeiros LJ, Chang K-C. Hodgkin lymphoma: a review of pathological features and recent advances in pathogenesis. Pathology. 2020;52(1):154-165. PubMed

4.Hasenclever D, Diehl V, Armitage JO, et al. A prognostic score for advanced Hodgkin's disease. N Engl J Med. 1998;339(21):1506-1514. PubMed

5.Straus DJ, Jung S-H, Pitcher B, et al. CALGB 50604: risk-adapted treatment of nonbulky early-stage Hodgkin lymphoma based on interim PET. Blood. 2018;132(10):1013-1021. PubMed

6.Sasse S, Bröckelmann PJ, Goergen H, et al. Long-term follow-up of contemporary treatment in early-stage Hodgkin lymphoma: updated analyses of the German Hodgkin study group HD7, HD8, HD10, and HD11 trials. J Clin Oncol. 2017;35(18):1999-2007. PubMed

7.Skoetz N, Will A, Monsef I, Brillant C, Engert A, von Tresckow B. Comparison of first‐line chemotherapy including escalated BEACOPP versus chemotherapy including ABVD for people with early unfavourable or advanced stage Hodgkin lymphoma. Cochrane Database Syst Rev. 2017(5).

8.Johnson P, Federico M, Kirkwood A, et al. Adapted treatment guided by interim PET-CT scan in advanced Hodgkin’s lymphoma. N Engl J Med. 2016;374(25):2419-2429. PubMed

9.Radford J, Illidge T, Counsell N, et al. Results of a trial of PET-directed therapy for early-stage Hodgkin’s lymphoma. N Engl J Med. 2015;372(17):1598-1607. PubMed

10.Borchmann P, Goergen H, Kobe C, et al. PET-guided treatment in patients with advanced-stage Hodgkin's lymphoma (HD18): final results of an open-label, international, randomised phase 3 trial by the German Hodgkin Study Group. The Lancet. 2017;390(10114):2790-2802. PubMed

11.Mauz-Körholz C, Metzger ML, Kelly KM, et al. Pediatric Hodgkin lymphoma. J Clin Oncol. 2015;33(27):2975-2985. PubMed

12.Castellino SM, LeBlanc ML, Herrera AF, et al. An intergroup collaboration for advanced stage classical Hodgkin lymphoma (cHL) in adolescents and young adults (AYA): SWOG S1826. J Clin Oncol. 2020;38(15_suppl):TPS8067-TPS8067.

13.Provencio M, Millán I, España P, et al. Analysis of competing risks of causes of death and their variation over different time periods in Hodgkin's disease. Clin Cancer Res. 2008;14(16):5300-5305. PubMed

14.Broccoli A, Zinzani PL. The role of transplantation in Hodgkin lymphoma. Br J Haematol. 2019;184(1):93-104. PubMed

15.Schmitz N, Pfistner B, Sextro M, et al. Aggressive conventional chemotherapy compared with high-dose chemotherapy with autologous haemopoietic stem-cell transplantation for relapsed chemosensitive Hodgkin's disease: a randomised trial. The Lancet. 2002;359(9323):2065-2071. PubMed

16.Linch DC, Goldstone AH, McMillan A, et al. Dose intensification with autologous bone-marrow transplantation in relapsed and resistant Hodgkin's disease: results of a BNLI randomised trial. The Lancet. 1993;341(8852):1051-1054. PubMed

17.Fermé C, Mounier N, Diviné M, et al. Intensive salvage therapy with high-dose chemotherapy for patients with advanced Hodgkin’s disease in relapse or failure after initial chemotherapy: results of the Groupe d’Études des Lymphomes de l’Adulte H89 trial. J Clin Oncol. 2002;20(2):467-475. PubMed

18.Stiff PJ, Unger JM, Forman SJ, et al. The value of augmented preparative regimens combined with an autologous bone marrow transplant for the management of relapsed or refractory hodgkin disease: A southwest oncology group phase II trial. Biol Blood Marrow Transplant. 2003;9(8):529-539. PubMed

19.Tarella C, Cuttica A, Vitolo U, et al. High-dose sequential chemotherapy and peripheral blood progenitor cell autografting in patients with refractory and/or recurrent Hodgkin lymphoma. Cancer. 2003;97(11):2748-2759. PubMed

20.Yuen AR, Rosenberg SA, Hoppe RT, Halpern JD, Horning SJ. Comparison between conventional salvage therapy and high-dose therapy with autografting for recurrent or refractory Hodgkin's disease. Blood. 1997;89(3):814-822. PubMed

21.Lavoie JC, Connors JM, Phillips GL, et al. High-dose chemotherapy and autologous stem cell transplantation for primary refractory or relapsed Hodgkin lymphoma: long-term outcome in the first 100 patients treated in Vancouver. Blood. 2005;106(4):1473-1478. PubMed

22.Longo DL, Duffey PL, Young RC, et al. Conventional-dose salvage combination chemotherapy in patients relapsing with Hodgkin's disease after combination chemotherapy: the low probability for cure. J Clin Oncol. 1992;10(2):210-218. PubMed

23.Mead G, Harker, G., Kushlan P and Rosenberg SA. Single agent palliative chemotherapy for end-stage hodgkin's disease. Cancer. 1982;50:829-835. PubMed

24.Proctor SJ, Lennard A.L., Jackson G.H, McKay P, Culligan D and Lucraft H.H. The role of an all-oral chemotherapy containing lomustine (CCNU) in advanced, progressive Hodgkin lymphoma: A patient-friendly palliative option which can result in long-term disease control. Ann Oncol. 2010;21(2):426-428. PubMed

25.Senter PD, Sievers EL. The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat Biotechnol. 2012;30(7):631-637. PubMed

26.Younes A, Gopal AK, Smith SE, et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin's lymphoma. J Clin Oncol. 2012;30(18):2183-2189. PubMed

27.Chen R, Gopal AK, Smith SE, et al. Five-year survival and durability results of brentuximab vedotin in patients with relapsed or refractory Hodgkin lymphoma. Blood. 2016;128(12):1562-1566. PubMed

28.Bröckelmann PJ, Zagadailov EA, Corman SL, et al. Brentuximab vedotin in patients with relapsed or refractory Hodgkin lymphoma who are Ineligible for autologous stem cell transplant: A Germany and United Kingdom retrospective study. Eur J Haematol. 2017;99(6):553-558. PubMed

29.Opdivo (nivolumab for injection): intravenous infusion, 10 mg nivolumab /mL 40 mg and 100 mg single-use vials [product monograph]. Montreal (QC): Bristol-Myers Squibb Canada Co.; 2021 May 21:1-2.

30.Timmerman JM, Engert A, Younes A, et al. Checkmate 205 update with minimum 12-month follow up: a phase 2 study of nivolumab in patients with relapsed/refractory classical Hodgkin lymphoma. Blood. 2016;128(22):1110-1110.

31.Ansell S, Gutierrez ME, Shipp MA, et al. A phase 1 study of nivolumab in combination with ipilimumab for relapsed or refractory hematologic malignancies (CheckMate 039). Blood. 2016;128(22):183-183.

32.Clinical study report: P204V02MK3475. A phase I/II study of pembrolizumab (MK-3475) in children with advanced melanoma or a PD-L1 positive advanced, relapsed or refractory solid tumor or lymphoma (KEYNOTE-051). [internal sponsor's report]. Kirkland (QC): Merck Canada Inc.; 2020 Apr 24.

33.Clinical study report: P087V02MK3475. A phase II clinical trial of MK-3475 (pembrolizumab) in subjects with relapsed or refractory (R/R) classical Hodgkin lymphoma (cHL). [internal sponsor's report]. Kenilworth (NJ): Merck Sharp & Dohme Corp.; 2020 Apr 9.

34.Clinical study report: P204V01MK3475. A phase III, randomized, open-label, clinical trial to compare pembrolizumab with brentuximab vedotin in subjects with relapsed or refractory classical Hodgkin lymphoma. [internal sponsor's report]. Kirkland (QC): Merck Canada Inc.; 2020 Apr 10.

35.CADTH pCODR Expert Review Committee (pERC) final recommendation: brentuximab vedotin (Adcetris). Ottawa (ON): CADTH; 2019 Mar 7: https://cadth.ca/sites/default/files/pcodr/Reviews2019/10145BrentuximabHL_FnRec_approvedbyChair_Post_07Mar2019_final.pdf. Accessed 2021 May 17.

36.Brockelmann PJ, Engert A. Checkpoint inhibition in Hodgkin lymphoma - a review. Oncol Res Treat. 2017;40(11):654-660. PubMed

37.Keytruda (pembrolizumab): powder for solution for infusion 50 mg solution for infusion 100 mg/4 mL vial antineoplastic agent, monoclonal antibody [product monograph]. Kirkland (QC): Merck Canada Inc.; 2021 Feb 5.

38.Adcetris (brentuximab vedotin for injection): lyophilized powder for reconstitution with 10.5 mL of sterile Water for Injection, USP 50 mg; Antineoplastic agent [product monograph]. Bothell (WA): Seattle Genetics, Inc.; 2013 Feb 1.

39.Fedorova LV, Lepik KV, Mikhailova NB, et al. Nivolumab discontinuation and retreatment in patients with relapsed or refractory Hodgkin lymphoma. Ann Hematol. 2021;100(3):691-698. PubMed

40.Shah GL, Moskowitz CH. Transplant strategies in relapsed/refractory Hodgkin lymphoma. Blood. 2018;131(15):1689-1697. PubMed

41.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

42.Grey matters: a practical tool for searching health-related grey literature. Ottawa (ON): CADTH; 2019: https://www.cadth.ca/grey-matters. Accessed 2021 Feb 3.

43.Chen R, Zinzani PL, Lee HJ, et al. Pembrolizumab in relapsed or refractory Hodgkin lymphoma: 2-year follow-up of KEYNOTE-087. Blood. 2019;134(14):1144-1153. PubMed

44.Cheson BD, Pfistner B, Juweid ME, et al. Revised response criteria for malignant lymphoma. J Clin Oncol. 2007;25(5):579-586. PubMed

45.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

46.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 May 17.

47.Pickard AS, Neary MP, Cella D. Estimation of minimally important differences in EQ-5D utility and VAS scores in cancer. Health Qual Life Outcomes. 2007;5:70. PubMed

48.Giesinger JM, Kieffer JM, Fayers PM, et al. Replication and validation of higher order models demonstrated that a summary score for the EORTC QLQ-C30 is robust. J Clin Epidemiol. 2016;69:79-88. PubMed

49.EORTC Quality of life: FAQs. Brussels (Belgium): EORTC Data Center: https://qol.eortc.org/faq/. Accessed 2021 Mar 8.

50.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

51.EORTC QLC-C30 scoring manual. Brussels (Belgium): EORTC Data Center; 2001: https://www.eortc.org/app/uploads/sites/2/2018/02/SCmanual.pdf. Accessed 2021 Mar 8.

52.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. Journal of Patient reported Outcomes. 2021;5(1):4. PubMed

53.Luo N, Fones CS, Lim SE, Xie F, Thumboo J, Li SC. The European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-c30): validation of English version in Singapore. Qual Life Res. 2005;14(4):1181-1186. PubMed

54.Snyder CF, Blackford AL, Sussman J, et al. Identifying changes in scores on the EORTC-QLQ-C30 representing a change in patients' supportive care needs. Qual Life Res. 2015;24(5):1207-1216. PubMed

55.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

56.Brooks R. EuroQol: the current state of play. Health Policy. 1996;37(1):53-72. PubMed

57.EuroQol Group. EuroQol--a new facility for the measurement of health-related quality of life. Health Policy. 1990;16(3):199-208. PubMed

58.Teckle P, Peacock S, McTaggart-Cowan H, et al. The ability of cancer-specific and generic preference-based instruments to discriminate across clinical and self-reported measures of cancer severities. Health Qual Life Outcomes. 2011;9:106. PubMed

Appendix 1: Clinical Literature Search Strategy

Note that this appendix has not been copy-edited.

Overview

Interface: Ovid

Databases:

Date of search: March 17, 2021

Alerts: Bi-weekly search updates until project completion

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

Limits:

Table 35: Syntax Guide

Syntax

Description

/

At the end of a phrase, searches the phrase as a subject heading

MeSH

Medical Subject Heading

exp

Explode a subject heading

*

Before a word, indicates that the marked subject heading is a primary topic; or, after a word, a truncation symbol (wildcard) to retrieve plurals or varying endings

#

Truncation symbol for 1 character

?

Truncation symbol for 1 or no characters only

adj#

Requires terms to be adjacent to each other within # number of words (in any order)

.ti

Title

.ot

Original title

.ab

Abstract

.hw

Heading word; usually includes subject headings and controlled vocabulary

.kf

Author keyword heading word (MEDLINE)

.kw

Author keyword (Embase)

.dq

Candidate term word (Embase)

.pt

Publication type

.rn

Registry number

.nm

Name of substance word (MEDLINE)

.yr

Publication year

medall

Ovid database code: MEDLINE All, 1946 to present, updated daily

oemezd

Ovid database code; Embase, 1974 to present, updated daily

Multi-Database Strategy

Search strategy:

  1. (Keytruda* or pembrolizumab* or lambrolizumab* or MK 3475 or MK3475 or Merck 3475 or HSDB 8257 or HSDB8257 or Sch 900475 or Sch900475 or DPT0O3T46P).ti,ab,ot,kf,hw,rn,nm.

  2. Hodgkin Disease/

  3. (Hodgkin* or reed sternberg*).ti,ab,kf,ot,hw.

  4. ((lymphoma* or lymphogranuloma* or granuloma*) adj5 malign*).ti,ab,kf,ot,hw.

  5. (classic* HL or classic* HD).ti,ab,kf,ot,hw.

  6. or/2-5

  7. 1 and 6

  8. 7 use medall

  9. *pembrolizumab/

  10. (Keytruda* or pembrolizumab* or lambrolizumab* or MK 3475 or MK3475 or Merck 3475 or HSDB 8257 or HSDB8257 or Sch 900475 or Sch900475).ti,ab,kw,dq.

  11. 9 or 10

  12. exp Hodgkin Disease/

  13. (Hodgkin* or reed Sternberg*).ti,ab,kw,dq.

  14. ((lymphoma* or lymphogranuloma* or granuloma*) adj5 malign*).ti,ab,kw,dq.

  15. (classic* HL or classic* HD).ti,ab,kw,dq.

  16. or/12-15

  17. 11 and 16

  18. 17 use oemezd

  19. 18 not (conference abstract or conference review).pt.

  20. 8 or 19

  21. remove duplicates from 20

Clinical Trials Registries

ClinicalTrials.gov

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

Search -- Studies with results on Keytruda (pembrolizumab) AND (Hodgkin disease OR Hodgkin lymphoma)

WHO ICTRP

International Clinical Trials Registry Platform, produced by the World Health Organization. Targeted search used to capture registered clinical trials.

Search terms – Keytruda (pembrolizumab) AND (Hodgkin disease OR Hodgkin lymphoma)

Health Canada’s Clinical Trials Database

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

Search terms – Keytruda (pembrolizumab) AND (Hodgkin disease OR Hodgkin lymphoma)

EU Clinical Trials Register

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

Search terms – Keytruda (pembrolizumab) AND (Hodgkin disease OR Hodgkin lymphoma)

Grey Literature

Search dates: March 12, 2021 – March 16, 2021

Keywords: Keytruda OR pembrolizumab OR MK-3475) AND (Hodgkin disease OR Hodgkin lymphoma)

Limits: No publication date limits

Updated: Search updated prior to the completion of stakeholder feedback period

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:

Appendix 2: Subsequent Anticancer Medication Utilization in KEYNOTE-204

Note that this appendix has not been copy-edited.

Table 36: Subsequent Anticancer Therapy in KEYNOTE-204

Anticancer therapy, n (%)

Pembrolizumab

(n = 148)

Brentuximab vedotin

(n = 152)

Total

104 (70.2)

148 (97.4)

MCL 1 inhibitor (unspecified)

0 (0)

1 (0.7)

Anti-CD25 antibody-drug conjugate

0 (0)

1 (0.7)

Anti-LAG-3 monoclonal antibody (unspecified) + anti-PD1 monoclonal antibody

1 (0.7)

0 (0)

Anti-LAG-3 monoclonal antibody (unspecified) + nivolumab

2 (1.4)

1 (0.7)

Anti-LAG-3 monoclonal antibody (unspecified) + pembrolizumab

1 (0.7)

0 (0)

Autologous redirected CD30 chimeric antigen receptor T-cells + fludarabine phosphate

1 (0.7)

0 (0)

Azacitidine

0 (0)

1 (0.7)

Bendamustine

1 (0.7)

5 (3.3)

Bendamustine + brentuximab vedotin

9 (6.1)

6 (3.9)

Bendamustine + brentuximab vedotin + dexamethasone

1 (0.7)

0 (0)

Bendamustine + brentuximab vedotin + doxorubicin

1 (0.7)

0 (0)

Bendamustine + carmustine + cytarabine + dexamethasone + etoposide + gemcitabine + melphalan

0 (0)

1 (0.7)

Bendamustine + dexamethasone + gemcitabine

1 (0.7)

2 (1.3)

Bendamustine + gemcitabine

0 (0)

1 (0.7)

Bendamustine + gemcitabine + vinorelbine

3 (2.0)

3 (2.0)

Bendamustine + gentamicin + vinorelbine

0 (0)

1 (0.7)

Bendamustine + lenalidomide

1 (0.7)

0 (0)

Bendamustine + methylprednisolone + prednisolone

0 (0)

1 (0.7)

Bendamustine + prednisolone

0 (0)

1 (0.7)

Bendamustine + rituximab

1 (0.7)

0 (0)

Bleomycin + cyclophosphamide + doxorubicin + etoposide + prednisolone + procarbazine + vincristine

1 (0.7)

0 (0)

Bleomycin + dacarbazine + doxorubicin + vinblastine

0 (0)

1 (0.7)

Bleomycin + dacarbazine + vinblastine

0 (0)

1 (0.7)

Brentuximab vedotin

37 (25.0)

7 (4.6)

Brentuximab vedotin + carboplatin + etoposide + ifosfamide

1 (0.7)

0 (0)

Brentuximab vedotin + cyclophosphamide + etoposide + prednisolone + procarbazine

0 (0)

1 (0.7)

Brentuximab vedotin + dexamethasone

1 (0.7)

0 (0)

Brentuximab vedotin + doxorubicin + gemcitabine + vinorelbine

2 (1.4)

0 (0)

Brentuximab vedotin + doxorubicin + etoposide + vinblastine

1 (0.7)

0 (0)

Brentuximab vedotin + gemcitabine + ifosfamide + prednisolone + vinorelbine

1 (0.7)

0 (0)

Brentuximab vedotin + nivolumab

0 (0)

1 (0.7)

Carboplatin + cisplatin + gemcitabine + prednisone

1 (0.7)

0 (0)

Carboplatin + etoposide + ifosfamide

0 (0)

3 (2.0)

Carmustine + cyclophosphamide + cytarabine + etoposide

1 (0.7)

0 (0)

Carmustine + cytarabine + dexamethasone + etoposide + melphalan

1 (0.7)

0 (0)

Chlorambucil

0 (0)

1 (0.7)

Chlorambucil + cyclophosphamide + methylprednisolone

0 (0)

1 (0.7)

Chlorambucil + dexamethasone + etoposide + lomustine

0 (0)

1 (0.7)

Cisplatin

1 (0.7)

0 (0)

Cisplatin + cytarabine + dexamethasone

1 (0.7)

5 (3.3)

Cisplatin + cytarabine + etoposide + methylprednisolone

1 (0.7)

2 (1.3)

Cisplatin + dexamethasone + gemcitabine

2 (1.4)

4 (2.6)

Cisplatin + gemcitabine

0 (0)

1 (0.7)

Cisplatin + gemcitabine + prednisone

0 (0)

1 (0.7)

Cyclophosphamide

2 (1.4)

1 (0.7)

Cyclophosphamide + cytarabine + etoposide

0 (0)

1 (0.7)

Cyclophosphamide + dexamethasone + procarbazine + vincristine

0 (0)

1 (0.7)

Cyclophosphamide + etoposide + prednisolone + procarbazine

0 (0)

2 (1.3)

Cyclophosphamide + etoposide + prednisone + procarbazine

1 (0.7)

0 (0)

Cyclophosphamide + methylprednisolone

0 (0)

1 (0.7)

Cyclophosphamide + prednisone + procarbazine + vincristine

2 (1.4)

0 (0)

Dacarbazine + doxorubicin + vinblastine

1 (0.7)

0 (0)

Dacarbazine + vinblastine

0 (0)

1 (0.7)

Dexamethasone

0 (0)

1 (0.7)

Dexamethasone + doxorubicin + gemcitabine + vinorelbine

0 (0)

1 (0.7)

Dexamethasone + mitoxantrone + vinblastine

0 (0)

1 (0.7)

Doxorubicin + vinblastine

1 (0.7)

0 (0)

Doxorubicin

1 (0.7)

0 (0)

Doxorubicin + etoposide + ifosfamide

0 (0)

1 (0.7)

Doxorubicin + gemcitabine + vinorelbine

0 (0)

1 (0.7)

Doxorubicin + pembrolizumab

0 (0)

1 (0.7)

Doxorubicin + vinblastine

0 (0)

1 (0.7)

Epirubicin + etoposide + ifosfamide

2 (1.4)

0 (0)

Etoposide

0 (0)

1 (0.7)

Etoposide + ifosfamide

1 (0.7)

0 (0)

Etoposide + ifosfamide + oxaliplatin

1 (0.7)

0 (0)

Gemcitabine

0 (0)

2 (1.3)

Gemcitabine + ifosfamide + vinorelbine

1 (0.7)

2 (1.3)

Gemcitabine + nivolumab

1 (0.7)

1 (0.7)

Gemcitabine + oxaliplatin

1 (0.7)

0 (0)

Gemcitabine + pembrolizumab + vinblastine

0 (0)

1 (0.7)

Gemcitabine + vinorelbine

2 (1.4)

2 (1.3)

Ibrutinib

0 (0)

1 (0.7)

Isatuximab

0 (0)

1 (0.7)

Methylprednisolone

0 (0)

1 (0.7)

Nivolumab

5 (3.4)

30 (19.7)

Nivolumab + signal regulatory protein alpha (recombinant) Fc fusion protein

1 (0.7)

0 (0)

Panobinostat

0 (0)

1 (0.7)

Pembrolizumab

2 (1.4)

27 (17.8)

Pembrolizumab + vinblastine

0 (0)

1 (0.7)

Pixantrone

0 (0)

1 (0.7)

Procarbazine + vinorelbine

1 (0.7)

0 (0)

Procarbazine

1 (0.7)

0 (0)

Rituximab

1 (0.7)

2 (1.3)

Vinblastine

1 (0.7)

5 (3.3)

Appendix 3: Description and Appraisal of Outcome Measures

Note that 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 minimal important difference (MID)):

Findings

Table 37: Summary of Outcome Measures and Their Measurement Properties

Outcome measure

Type

Conclusions about measurement properties

MID

EORTC QLQ-C30

30-item, patient-reported, cancer-specific, quality of life questionnaire using 4- and 7-point Likert scales.

Validity, Reliability, and Responsiveness:

  • Evidence of validity and reliability in populations with cancer. No literature was identified that assessed responsiveness in populations with cancer.

  • No evidence supporting adequate validity, reliability, and responsiveness in patients with relapsed or refractory cHL.

Patients with cancer:

  • 5-10 points small

  • 10-20 points moderate

  • > 20 points large clinical change

  • 9-32 points for improvement

  • 7-21 points for deterioration

No MID specific to relapsed or refractory cHL

EQ-5D-3L

Patient-reported, generic quality of life instrument using a 3-point ordinal scale to assess health in 5 dimensions.

Validity: Moderate to poor ability to distinguish between cancer severity by 3 scales (self-reported health status, ECOG-PS, stage of cancer).

Reliability: No literature was identified that assessed responsiveness in patients with relapsed or refractory cHL.

Responsiveness: No literature was identified that assessed responsiveness in patients with relapsed or refractory cHL.

MID 0.033-0.074 estimated for the general population.

MID 0.07-0.11 for UK-index scores and 0.05-0.08 for US-index scores for patients with cancer.

Visual analogue scale MID 6-10 for patients with cancer.

No MID identified in patients with relapsed or refractory cHL.

EORTC QLQ-C30 = European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire – core 30 items; EQ-5D-3L = European Quality of Life Scale – 5 Dimensions – 3 Levels; MID = minimal important difference.

EORTC QLQ-C30

Description

The European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Core 30, or EORTC QLQ-C30, is 1 of the most commonly used patient-reported outcome measures in oncology clinical trials.48 It is a multi-dimensional, cancer-specific, evaluative measure of HRQoL. It was designed specifically for the purpose of assessing changes in participants’ HRQoL in clinical trials in response to treatment.49 The core questionnaire of the EORTC QLQ-C30 consists of 30 questions that are scored to create 5 multi-item functional scales, 3 multi-item symptom scales, 6 single-item symptom scales, and a 2-item quality of life (QoL) scale, as outlined in Table 25. The first 2 versions of the questionnaire have been previously validated in patients with cancer.50 Version 3.0 of the questionnaire is the most current version and has been in use since December of 1997.51 It is available in 90 languages and is intended for use in adult populations only. The global QoL scale is also known as the Global Health Status (GHS).52

Table 38: EORTC QLQ-C30 Scales

Functional scales

(15 questions)

Symptom scales

(7 questions)

Single-item symptom scales

(6 questions)

Global quality of life

(2 questions)

Physical function (5)

Role function (2)

Cognitive function (2)

Emotional function (4)

Social function (2)

Fatigue (3)

Pain (2)

Nausea and vomiting (2)

Dyspnea (1)

Insomnia (1)

Appetite loss (1)

Constipation (1)

Diarrhea (1)

Financial impact (1)

Global Quality of Life (2)

Scoring

The EORTC QLQ-C30 uses a 1-week recall period to assess function and symptoms.51 Most questions have 4 response options (“not at all,” “a little,” “quite a bit,” “very much”), with scores on these items ranging from 1 to 4. For the 2 items that form the global QoL scale, the response format is a 7-point Likert-type 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.51 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 QoL scale would reflect an improvement. According to the EORTC QLQ-C30 scoring algorithm, if there are missing items for a scale (i.e., the participant 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.

Validity

One cross-sectional study aimed to validate the EORTC QLQ-C30 in a convenience sample of cancer patients in Singapore.53 Most patients had breast and colorectal cancers, but leukemia, lung cancer, lymphoma, germ cell tumour, and other cancers were also reported. Construct validity was assessed by cross-sectional correlational evidence and discriminative evidence. First, convergent validity was assessed using Spearman’s correlations between QLQ-C30 and Short Form-36 (SF-36) scales, hypothesizing moderate to strong correlation (defined as correlation coefficient of 0.35 to 0.5, and > 0.5, respectively) between scales of these 2 instruments measuring similar dimensions of HRQoL. Results showed moderate to strong correlations between QLQ-C30 and SF-36 scales, ranging from 0.35 to 0.67 across the assessed scales. Next, known-groups approach was used to compare 6 QLQ-C30 scale scores between patients reporting mild and severe symptoms, as well as by stage of disease and presence of comorbid conditions. Except for emotional functioning, the remaining 5 scales showed better scores in patients with mild symptoms than those with severe symptoms (P < 0.05 for all other comparisons). Patients in early stages of cancer (or with no comorbid conditions) generally had better QLQ-C30 scores than those in advanced disease stages (or with comorbid conditions); however, none of these differences were statistically significant.

A recent cross-sectional study in Kenya was conducted to evaluate the psychometric properties of the EORTC QLQ-C30, using the English or Kiswahili version in 100 patients with cancer.52 Most patients had breast cancer, followed by prostate, Kaposi sarcoma, lung, and other cancers. Construct validity was assessed by examining the inter-scale correlations among the subscales of EORTC QLQ-C30. The inter-scale correlations were weak to strong with an absolute magnitude ranging from 0.07 to 0.73. Notably, apart from cognitive functioning, emotional functioning, nausea and vomiting, dyspnea, appetite loss, constipation, and diarrhea, the GHS correlated moderately with the remaining subscales (r ≥ 0.30). Cross-cultural validity was evaluated but not reported here.

Reliability

The Singaporean cross-sectional study above also assessed internal consistency reliability by calculating Cronbach alpha for all QLQ-C30 scales.53 Cronbach alpha was ≥ 0.70 for 6 of the 9 assessed QLQ-C30 scales; cognitive functioning, physical functioning, and nausea and vomiting had a Cronbach alpha ranging from 0.19 to 0.68. The Kenyan study described above assessed the internal consistency of each scale of the questionnaire using Cronbach alpha coefficients.52 With the exception of the cognitive function scale, all of the scales had a Cronbach alpha ≥ 0.70.

No studies evaluating the responsiveness of the instrument were found.

Minimum Important Difference

For use in clinical trials, scores on the EORTC QLQ-C30 can be compared between different groups of patients or within a group of patients over time. One study from 1998 conducted in patients with breast cancer and small cell lung cancer estimated a clinically relevant change in score on any scale of the EORTC QLQ-C30 to be 10 points.45 The estimate was based on a study that used an anchor-based approach to estimate the minimum important difference in which patients who reported “a little” change (for better or worse) on the subjective significance questionnaire had corresponding changes on a function or symptom scale of the EORTC QLQ-C30 of approximately 5 to 10 points. Participants 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.

More recently in 2015, a Canadian study estimated the MIDs of EORTC QLQ-C30 scales using data from 193 patients newly diagnosed with breast and colorectal cancers.54 The Supportive Care Needs Survey-Short Form-34 (SCNS-SF34) was used as an anchor; mean changes in EORTC QLQ-C30 scales associated with improvement, worsening, and no-change in supportive care based on the SCNS-SF34 was then calculated. MIDs were assessed for the following scales: physical function, role function, emotional function, global health/QoL (i.e., GHS), pain, and fatigue. For improvement, MIDs associated with a statistically significant improvement in supportive care needs ranged from 10 to 32 points. For worsening, MIDs associated with a statistically significant worsening of supportive care needs ranged from 9 to 21 points. The range for unchanged supportive care needs was from 1-point worsening to 16-point improvement in EORTC QLQ-C30 score. Based on this, the authors suggested a 10-point change in EORTC QLQ-C30 score represented changes in supportive care needs, and therefore, should be considered for clinical use.

In 2014, another Canadian study estimated the MID for EORTC QLQ-C30 in 369 patients with advanced cancer who completed the questionnaire at baseline and 1 month after radiation.55 The most common cancer type was breast cancer, followed by lung, prostate, gastrointestinal, renal cell, and other cancers. MID was estimated using both anchor- and distribution-based methods for improvement and deterioration. Two anchors of overall health and overall QoL were used, both taken directly from the EORTC QLQ-C30 (questions 29 and 30) where patients rated their overall health and QoL 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-3L

The European Quality of Life Scale (EQ-5D) is a generic HRQoL instrument that may be applied to a wide range of health conditions and treatments.56,57 The first of 2 parts of the EQ-5D is 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-3L has 3 possible levels (1, 2, or 3) for each domain representing ‘no problems,’ ‘some problems,’ and ‘extreme problems,’ respectively. Respondents are asked to choose the level that reflects their health state for each of the 5 dimensions, corresponding with 243 different health states. A scoring function can be used to assign a value (EQ-5D-3L index score) to self-reported health states from a set of population-based preference weights.56,57 The second 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. Hence, the EQ-5D 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, such as 11121, 33211, and so forth,

  2. A population preference-weighted health index score based on the descriptive system,

  3. A self-reported assessment of health status based on the EQ-VAS.

The EQ-5D index score is generated by applying a multi-attribute utility function to the descriptive system. Different utility functions are available that reflect the preferences of specific populations (e.g., US or UK). The lowest possible overall score for the 3L version (corresponding to severe problems on all 5 attributes) varies depending on the utility function that is applied to the descriptive system (e.g., −0.59 for the UK algorithm and −0.109 for the US algorithm). 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. Reported MIDs for the 3L version of the scale have ranged from 0.033 to 0.074.46

Teckle et al. conducted a study of patients (n=184) who had either breast (36%), colorectal (31%), or lung (33%) cancer at the Vancouver Cancer Clinic to investigate if disease severity could be distinguished by cancer-specific and generic preference-based instruments.58 Internal consistency was calculated using Cronbach alpha and all 5 functioning scales along with GHS showed acceptable consistency (α > 0.7) with values ranging from 0.77 to 0.82. Validity was assessed using Pearson’s correlation coefficient (r) where r between 0 and 0.3 demonstrated weak correlation, between 0.3 and 0.49 was moderate, and greater than 0.5 was considered strong. Teckle et al. found the following, between the EORTC QLQ-C30, and EQ-5D, r = 0.43; comparing the EORTC QLQ-C30 and EQ-VAS, r = 0.73; and between EQ-5D and EQ-VAS, r = 0.43. External validity was estimated between cancer severity (self-reported health status, Eastern Cooperative Oncology Group-Performance Status [ECOG-PS], and cancer stage). An effect size (ES) between 0.2 and 0.5 was considered small, between 0.5 and 0.8 was medium, and greater than 0.8 was large. The EQ-5D was able to discriminate populations based on self-reported health status (excellent/good versus fair/very poor; ES = 0.90), and somewhat based on ECOG-PS (0 versus 1 to 3; ES = 0.31), but not for stage of cancer (stages 1 and 2 versus stages 3 and 4; ES = 0.06). The EORTC QLQ-C30 performed better in all 3 areas: self-reported health status (ES = 1.39), ECOG-PS (ES = 0.65), and stage of cancer (ES = 0.49). It is worth noting that the EQ-5D was based on a non-Canadian population and the comparison with EORTC QLQ-C30 was based solely on the 2 questions asking about overall health and HRQoL rather than the questionnaire as a whole. This study was a mixed population of 3 types of cancer and the results may not exactly reflect what would be observed in patients with relapsed or refractory cHL. Furthermore, there was no information on what type of treatment the patients were receiving when completing the questionnaires.

Pickard et al. conducted a retrospective analysis of 534 patients with 11 types of cancer (including colon/rectal cancer) to estimate the MID using distribution-based (SEM, 1/2 SD, and 1/3 SD) and anchor-based (ECOG) methods.47 After stratifying by ECOG status, the mean weighted index score MID for all cancer patients was estimated to be between 0.07 and 0.11 for UK-index scores and between 0.05 and 0.08 for US-index scores. The VAS MID was estimated to range from 6 to 11 points for all patients. No MID information was identified in patients with relapsed or refractory classical Hodgkin’s Lymphoma.

Appendix 4: PFS Survival Curves for KEYNOTE-051 and KEYNOTE-087 and OS Survival Curves for KEYNOTE-087

Figure 5: Kaplan-Meier Estimates of PFS in KEYNOTE-051

A Kaplan-Meier progression-free survival curve for 7 eligible patients treated in the Keynote-051 trial from 0 to 16 months. The curve starts descending at 2 months with the number of at-risk patients at 0, 2, 4, 6, 8, 10, 12, and 14 months being 7, 3, 2, 2, 2, 1, 1, and 0, respectively.

Figure 6: Kaplan-Meier Estimates of PFS in KEYNOTE-087, Cohort 1

A Kaplan-Meier progression-free survival curve for patients in cohort 1 of the KEYNOTE-087 trial, from 0 to 40 months. The number of patients at risk are shown at the beginning of each 3-month time period.

Figure 7: Kaplan-Meier Estimates of PFS in KEYNOTE-087, Cohort 2

A Kaplan-Meier progression-free survival curve for patients in cohort 2 of the KEYNOTE-087 trial from 0 to 39 months. The number of patients at risk are shown at the beginning of each 3-month time period.

Figure 8: Kaplan-Meier Estimates of PFS in KEYNOTE-087, Cohort 3

A Kaplan-Meier progression-free survival curve for patients in cohort 3 of the KEYNOTE-087 trial, from 0 to 42 months. the number of patients at risk are shown at the beginning of each 3-month time period.

Figure 9: Kaplan-Meier Estimates of Overall Survival in KEYNOTE-087, Cohort 1

A Kaplan-Meier overall survival curve for patients in cohort 1 of the KEYNOTE-087 trial from 0 to 44 months. The number of patients at risk are shown at the beginning of each 3-month time period. Median OS is not reached, with the proportion of patients with no death event being greater than 80% at 44 months.

Figure 10: Kaplan-Meier Estimates of Overall Survival in KEYNOTE-087, Cohort 2

A Kaplan-Meier overall survival curve for patients in cohort 2 of the KEYNOTE-087 trial from 0 to 44 months. The number of patients at risk are shown at the beginning of each 3-month time period. Median OS is not reached, with the proportion of patients with no death event being greater than 80% at 44 months.

Figure 11: Kaplan-Meier Estimates of Overall Survival in KEYNOTE-087, Cohort 3

A Kaplan-Meier overall survival curve for patients in cohort 3 of the KEYNOTE-087 trial from 0 to 45 months. The number of patients at risk are shown at the beginning of each 3-month time period. Median OS is not reached, with the proportion of patients with no death event being around 80% at 45 months.

Pharmacoeconomic Review

Abbreviations

ASCT

autologous stem cell transplantation

BIA

budget impact analysis

BV

brentuximab vedotin

cHL

classic Hodgkin lymphoma

ICER

incremental cost-effectiveness ratio

OS

overall survival

pCODR

CADTH pan-Canadian Oncology Drug Review

PFS

progression-free survival

QALY

quality-adjusted life-year

SCT

stem cell transplant

WTP

willingness to pay

Executive Summary

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

Table 1: Submitted for Review

Item

Description

Drug product

Pembrolizumab (Keytruda), 100 mg per 4 mL vial of solution for IV infusion

Submitted price

Pembrolizumab 100 mg: $4,400.00

Indication

Adult and pediatric patients with refractory or relapsed classical Hodgkin lymphoma, as monotherapy, who have failed ASCT, or who are not candidates for multi-agent salvage chemotherapy and ASCT

Health Canada approval status

NOC

Health Canada review pathway

Other expedited pathway: Project Orbis

NOC date

February 5, 2021

Reimbursement request

As per indication

Sponsor

Merck Canada Inc.

Submission history

Previously reviewed: Yes

Indication: classical Hodgkin lymphoma

Recommendation date: January 5, 2018

Recommendation: Reimburse with clinical criteria and/or conditions

ASCT = autologous stem cell transplant; NOC = Notice of Compliance.

Table 2: Summary of Economic Evaluation

Component

Description

Type of economic evaluation

Cost-utility analysis

Partition survival model

Target populations

  • Adult patients with refractory or relapsed classical Hodgkin lymphoma who relapsed post-ASCT or were ineligible for ASCT

  • Pediatric patients with refractory or relapsed classical Hodgkin lymphoma who relapsed post-ASCT or were ineligible for ASCT

Treatment

Pembrolizumab

Comparator

BV

Perspective

Canadian publicly funded health care payer

Outcome

QALYs, LYs

Time horizon

35 years

Key data source

  • KEYNOTE-204 trial to inform PFS

  • Published literature to inform overall survival

Submitted results

  • Adult population: Pembrolizumab is dominant (incremental cost savings $24,231; incremental QALYs gained 0.840)

  • Pediatric population: Pembrolizumab is dominant (incremental cost savings $47,937; incremental QALYs gained 0.843)

Key limitations

  • The sponsor, in their base case, evaluated pembrolizumab against BV in a mixed population consisting of individuals who were either ASCT eligible (and relapsed) or ineligible. Across Canada, BV has limited utilization in an ASCT-ineligible subpopulation and was not recommended by the CADTH pan-Canadian oncology drug review in this patient population.

  • Chemotherapy is primarily used in the treatment of patients who are ASCT ineligible. Given a lack of direct or indirect comparative evidence, the cost-effectiveness of pembrolizumab vs. chemotherapy, and therefore, the cost-effectiveness in an ASCT-ineligible population, could not be determined.

  • Treatment-specific utilities values were used by the sponsor in their model, which overestimates long-term QALY gains associated with pembrolizumab. The utility benefit for pembrolizumab during the trial was applied across the 35-year time horizon, thus assuming that even post-treatment, pembrolizumab provides an indefinite utility benefit.

  • The sponsor assumed the observed PFS benefits for pembrolizumab in the trial continued past the trial duration. Clinical experts consulted by CADTH for this review anticipated that any benefit from pembrolizumab over existing therapies would likely be negligible after 10 years; therefore, treatment waning needed to be incorporated in long-term extrapolations of PFS.

  • The sponsor used data which did not censor individuals who received an SCT after primary treatment initiation, thus capturing the benefit of an SCT in PFS estimates. This approach was deemed inappropriate given insignificant but differential SCT rates between treatments, and that its inclusion artificially inflates PFS extrapolation across the model time horizon.

  • Subsequent therapy use did not align with what was seen in the trial.

  • Drug wastage was not incorporated in the sponsor’s base case; however, the product monograph for pembrolizumab states it is supplied as a single-use vial, and weight-based dosing is used in the pediatric population.

  • The sponsor’s model was unnecessarily complex and lacked transparency, preventing CADTH from fully validating the model and its findings. CADTH identified some errors in the model coding.

CADTH reanalysis results

  • In the CADTH reanalysis, only patients who are ASCT eligible were evaluated, as the cost-effectiveness of pembrolizumab in patients who are ASCT ineligible could not be determined given a lack of comparative evidence on pembrolizumab vs. chemotherapy.

  • Changes to derive a CADTH base case included: only evaluating an ASCT-eligible population; adding health state-specific utility values; incorporating treatment waning; using PFS data that censors SCT events; including drug wastage; and using Canadian gemcitabine costs.

  • Based on the CADTH reanalysis in patients who are ASCT eligible, pembrolizumab compared with BV is:

    • Dominant (less costly and more effective) in a pediatric population

    • Associated with an ICER of $733,624 per QALY gained in an adult population

  • A price reduction of 13% is required for pembrolizumab to be considered cost-effective at a WTP threshold of $50,000 per QALY in an adult ASCT-eligible population.

  • Using an alternative breakdown of subsequent therapy use from the trial, the ICER for pembrolizumab compared with BV increased to $532,115 in pediatrics and $2,071,825 in adults. A 29% price reduction is needed to ensure cost-effectiveness at a $50,000 per QALY threshold.

  • The cost-effectiveness of pembrolizumab compared with chemotherapy in patients who are ASCT ineligible is unknown.

ASCT = autologous stem cell transplant; BV = brentuximab vedotin; ICER = incremental cost-effectiveness ratio; LY = life-year; PFS = progression-free survival; QALY = quality-adjusted life-year; SCT = stem cell transplant; vs. = versus; WTP = willingness to pay.

Conclusions

Evidence suggests pembrolizumab provides statistically and clinically significant improvements in progression-free survival (PFS) |||||||||||||||||||||||||||||||||||| ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||. Across all studies, brentuximab vedotin (BV) patients were generally less likely to experience adverse events but more likely to discontinue therapy due to an adverse event. No comparators beyond BV were evaluated. Therefore, the comparative evidence of pembrolizumab to chemotherapy in an autologous stem cell transplant (ASCT)-ineligible subpopulation is unknown, due to a lack of direct or indirect evidence for PFS and overall survival (OS).

CADTH addressed several key limitations of the sponsor’s model by selecting appropriate comparators for the Canadian context; using PFS data that censored upon stem cell transplant (SCT) events; considering a treatment effect waning; changing treatment-specific utilities to disease-specific; and incorporating drug wastage. According to CADTH’s base case for ASCT-eligible adult patients, pembrolizumab is associated with an incremental cost-effectiveness ratio (ICER) of $733,624 per quality-adjusted life-year (QALY) gained when compared with BV, and in an ASCT-eligible pediatric population pembrolizumab is dominant (i.e., cost saving and associated with more QALYs). In adults, a price reduction of approximately 13% is required for pembrolizumab to be cost-effective at a willingness-to-pay (WTP) threshold of $50,000 per QALY. This result is contingent on pembrolizumab generating approximately $43,000 of cost savings due to patients who receive pembrolizumab first line not receiving immunotherapies such as nivolumab and pembrolizumab second line. There is a significant amount of outstanding uncertainty regarding the degree of these cost savings. A scenario analysis that altered the subsequent therapy distribution based on data from the KEYNOTE-204 trial found the ICER in adults increased to $2,071,825 per QALY. At this level, a 29% price reduction is needed to achieve a $50,000 per QALY threshold.

The conclusions of CADTH’s analysis are specific to an ASCT-eligible population. CADTH was unable to assess the cost-effectiveness of pembrolizumab in an ASCT-ineligible population due to an absence of comparative evidence between pembrolizumab and chemotherapy. In Canada, individuals who are ineligible for ASCT rarely receive BV; therefore, chemotherapy is the most relevant comparator. Drug costs for pembrolizumab are substantially higher relative to chemotherapy, as such information on this comparison is needed to assess the value of pembrolizumab in this subgroup. Finally, results for the pediatric population are entirely extrapolated from the adult data, so any uncertainty with that extrapolation will impact the robustness of the pediatric results.

Stakeholder Input Relevant to the Economic Review

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

One patient group, Lymphoma Canada, conducted 2 online surveys of patients with Hodgkin lymphoma. The first was conducted in 2017 from June 5 to 30, 2017, and 91 participants responded, while the second collected responses from November 6, 2020, to January 13, 2021, and 37 responded. Of the participants who provided demographic information, 55% resided in Canada. Of the 9 participants with pembrolizumab experience, 7 resided in Canada. All 9 participants had at least 2 prior lines of conventional chemotherapy before initiating pembrolizumab. Those previous chemotherapy treatments included 8 reports of ABVD (Adriamycin-bleomycin-vinblastine-dacarbazine), 6 reports of GDP (gemcitabine-dexamethasone-cisplatin), 2 reports of COPP (cyclophosphamide-Oncovin-procarbazine-prednisone), and 1 report for either DHAP (dexamethasone-cytarabine-cisplatin), bendamustine, or Revlimid. The reason for starting pembrolizumab included no other treatment options available (n = 2), progression after ASCT and a desire not to risk toxicity of an allogeneic SCT (n = 4), hoping for remission to proceed to allogeneic transplant (n = 1), and lack of response to 3 previous chemotherapy lines and no desire to undergo ASCT (n = 2). Eight of the pembrolizumab patients tolerated the therapy well, while the remaining patient stopped treatment due to toxicity and side effects (i.e., neuropathy and inflammatory arthritis).

Clinician input was received from Ontario Health (Cancer Care Ontario) Hematology Disease Site Drug Advisory Committee, Lymphoma Canada Scientific Advisory Board, and the Pediatric Oncology Group of Ontario. For adult patients, the current pathway of care was divided into whether patients were eligible or ineligible for ASCT. In those eligible and who failed ASCT, BV is the standard of care, while in those not eligible there is no clear gold standard, and a variety of approaches can be used including combination chemotherapy, radiation, clinical trials, or occasionally novel agents such as BV or anti-PD1 antibodies (pembrolizumab or nivolumab). The introduction of pembrolizumab would replace BV as monotherapy in those who failed ASCT, and in those ineligible for ASCT it provides a therapy to patients with limited treatment options. For pediatric patients, the vast majority are eligible for ASCT. In pediatric patients who fail ASCT, pembrolizumab is expected to serve as an additional line of therapy with the aim to provide a curative therapeutic option.

Feedback from drug plans indicated that, in the subpopulation of adult patients with classic Hodgkin lymphoma (cHL) who are ASCT ineligible, the comparator is palliative care and not BV (due to its limited funding). In the pediatric population, they noted that there does not appear to be a standard of care, but most patients were ASCT eligible. The plans also noted drug wastage could be minimized by vial sharing, but it likely is not feasible in small outpatient cancer centres.

The following stakeholder input was addressed in the sponsor’s models:

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

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

Economic Review

The current review is for pembrolizumab (Keytruda) for adult and pediatric patients with refractory or relapsed cHL who have failed ASCT or who are not candidates for multi-agent salvage chemotherapy and ASCT.

Economic Evaluation

Summary of Sponsor’s Economic Evaluation

Overview

The sponsor submitted a cost-utility analysis assessing pembrolizumab compared to BV in 2 distinct populations of adults or pediatric patients, with refractory or relapsed cHL who have failed ASCT or who are not candidates for multi-agent salvage chemotherapy and ASCT.1 The modelled population aligned with the Health Canada indication and the sponsor’s reimbursement request.

Pembrolizumab is a monotherapy available as a solution (100 mg/4 mL [25 mg/mL]) for IV infusion. The recommended dose of pembrolizumab in adult patients is 200 mg and 2 mg/kg (up to a maximum of 200 mg) in pediatric patients. Pembrolizumab is administered as an IV infusion for 30 minutes every 3 weeks until disease progression or unacceptable toxicity, or up to 24 months in patients without disease progression.2 The cost for pembrolizumab is $4,400 per 100 mg vial, equating to a cost per 21-day cycle of $8,800.00 in adults and $5,148.00 in a pediatric population (assuming no wastage). Pembrolizumab was compared to BV, which has a cost per 21-day cycle of $13,320.64 in adults and $10,193.04 in a pediatric population.

The clinical outcomes were QALYs and life-years, which were modelled over a 35-year time horizon. The base-case analysis was conducted from the Canadian publicly funded health care payer perspective, and a 1.5% discount rate was applied to both costs and outcomes.

Model Structure

The sponsor submitted a 3-state partitioned survival model with 3 mutually exclusive states consisting of “progression free,” “progressed disease,” and “death.” Membership of each state at any point in time is based on direct modelling of OS and PFS curves, which the sponsor extrapolated over the time horizon of the analysis. A piecewise approach was used to extrapolate Kaplan-Meier data, which consists of using Kaplan-Meier data until a set cut-off point (i.e., 52 weeks for PFS and time on treatment, and 135 weeks for OS), then fitting a parametric curves to the remaining data to extrapolate to the time horizon. During the model, the patient may discontinue treatment, at which point the cost of treatment is no longer incurred.

The cycle length is 1 week, with no correction made for events occurring within a cycle (i.e., half-cycle correction). The sponsor provided a graphical representation of the model which has been reproduced in Appendix 3, Figure 1.

Model Inputs

For the adult population, baseline characteristics of the target populations aligned with the KEYNOTE-204 trial, while the KEYNOTE-051 trial was used for the pediatric population.3,4 The mean ages for the adult and pediatric populations were 41.35 years and 14.90 years, respectively. Acquisition costs for weight-dependent or surface area–dependent therapies were calculated using the mean weight and body surface area of both the adult (weight: 76.45 kg; total body surface area: 1.90 m2) and pediatric populations (weight: 58.5 kg; total body surface area: 1.60 m2).

The KEYNOTE-204 trial was the primary source of efficacy data for the model.3 Treatment efficacy was modelled in terms of delaying time to progression and extending OS. PFS for both the adult and pediatric population were extrapolated from the KEYNOTE-204 trial data for both pembrolizumab and BV. The sponsor indicated PFS data from KEYNOTE-051 was not used for the pediatric population due to its small sample size of 7 patients with relapsed or refractory Hodgkin lymphoma.4 The other treatment efficacy outcome, OS, was not pulled from either of the previously mentioned KEYNOTE studies due to a lack of OS end point data. Therefore, it was derived from a phase II trial of BV in patients with relapsed or refractory Hodgkin lymphoma after failed ASCT.5 By using this data, the sponsor assumed no difference in OS between BV and pembrolizumab, and that the OS of those who failed ASCT was equivalent to the population ineligible for multi-agent salvage chemotherapy and ASCT. In addition to PFS and OS, time on treatment was extrapolated from the KEYNOTE-204 and KEYNOTE-051 trials for the adult and pediatric populations, respectively.3,4

For each treatment, Kaplan-Meier data were used until a user defined cut-off point (52 weeks for PFS and time on treatment and 135 weeks for OS), after which parametric survival curves were fitted to and extrapolated up to the time horizon. Seven different parametric approaches were considered, reflecting exponential, Weibull, log-normal, log-logistic, Gompertz, gamma, and generalized gamma distributions. The sponsor determined the “best-fitting” parametric distributions using statistical tests based on the Akaike Information Criterion and the Bayesian information criterion, combined with visual inspection. The sponsor also reported that clinical appropriateness was considered in selecting final distribution functions for the model. Based on these criteria, the sponsor adopted log-normal extrapolations for PFS, time on treatment, and OS in the adult and pediatric populations. The model allowed for consideration of “treatment effect waning,” but this was not considered by the sponsor in their base-case analysis.

In the sponsor’s base case, chemotherapy was not included as a comparator. The sponsor stated there was a lack of evidence to complete an analysis against pembrolizumab (in both the population who failed ASCT and who were ineligible for multi-agent salvage chemotherapy and ASCT), due to either insufficient outcome reporting, unrepresentative target population, or insufficient sample size. However, chemotherapy was included in a scenario analysis. In that scenario, parameters for time on treatment and PFS were assumed equal to BV, and the sponsor assumed a median OS of 36 months and extrapolated an OS curve using an exponential equation.

The dosing used in the model is consistent with that described in the Overview, where the recommended dose of pembrolizumab in adult patients is 200 mg and 2 mg/kg (up to a maximum of 200 mg) in pediatric patients. The reference case analysis assumed no drug wastage in the adult and pediatric populations using single-use vials of pembrolizumab.

Utility values were modelled based on EuroQol 5-Dimensions 3-Levels questionnaire data collected in the KEYNOTE-204 trial, mapped to the US tariff.3 Mean utility values for pembrolizumab and BV are 0.883 and 0.822 for “progression free,” and 0.861 and 0.766 for “progressed disease,” respectively. In scenario analyses, a mean disutility decrement of 0.075 is applied for grade 3 to 5 treatment-related adverse events identified in the KEYNOTE-204 trial.

The economic model included costs for drug acquisition, drug administration, subsequent treatment lines, SCTs, disease management by health state, terminal care (i.e., last 3 months before death), and adverse events. Drug acquisition costs were pulled from previous CADTH pan-Canadian Oncology Drug Review (pCODR) reports,6,7 while costs for SCTs were sourced from a cost-effectiveness analysis on chronic myeloid leukemia and data from the Canadian Institute for Health Information.8 Disease management costs were sourced from an economic evaluation on BV,9 and adverse events were derived from either the Canadian Institute for Health Information Patient Cost Estimator, Ontario Case Costing Initiative, or sponsor assumptions.

Summary of Sponsor’s Economic Evaluation Results

The results of the sponsor’s base case are presented below. The base case used a probabilistic analysis with 5,000 iterations, and scenario analyses were conducted in the adult and pediatric populations which disaggregated results into those eligible and ineligible for ASCT. Additional details pertaining to the sponsor’s submission are available in Appendix 3.

Base-Case Results

In the sponsor’s base-case analysis for the adult population, pembrolizumab was associated with an expected cost savings of $24,230 and a gain of 0.8408 QALYs over a 35-year time horizon (Table 3). As treatment with BV was more costly and produced fewer QALYs, the ICER of pembrolizumab compared to BV indicated pembrolizumab was dominant (less costly and more effective) in the target population (adult patients with refractory or relapsed cHL who have failed ASCT or who are not candidates for multi-agent salvage chemotherapy and ASCT). The sponsor reported that at a WTP threshold of $50,000 per QALY, the probability of pembrolizumab being cost-effective was 67% (note: errors were found in the cost-effectiveness acceptability curve resulting in the curve not being updated for pairwise comparisons; therefore, the sponsor-reported probabilities are uncertain).

Most of the total QALYs for each strategy were generated after the trial period (80%), with the remaining QALYs generated within the trial period (20%). Key savings in the total average costs for pembrolizumab included progressed disease costs (–$58,289) and subsequent treatment costs (–$47,243); while higher costs were primarily found for progression-free costs ($28,254) and acquisition costs ($52,976).

Table 3: Summary of the Sponsor’s Economic Evaluation Results (Adult Population)

Drug

Total costs ($)

Incremental costs ($)

Total QALYs

Incremental QALYs

ICER vs. BV

($/QALY)

BV

418,980

Reference

7.1236

Reference

Reference

Pembrolizumab

394,750

–24,230

7.9644

0.8408

Dominant

BV = brentuximab vedotin; ICER = incremental cost-effectiveness ratio; QALY = quality-adjusted life-year; vs. = versus.

Source: Sponsor’s pharmacoeconomic submission.1

In the sponsor’s base-case analysis for the pediatric population, pembrolizumab was associated with an expected cost savings of $47,937 and a gain of 0.8437 QALYs over a 35-year time horizon (Table 4). Similar to the results in the adult population, pembrolizumab was dominant over BV. At a WTP threshold of $50,000 per QALY, the probability of pembrolizumab being cost-effective was 67%.

Table 4: Summary of the Sponsor’s Economic Evaluation Results (Pediatric Population)

Drug

Total costs ($)

Incremental costs ($)

Total QALYs

Incremental QALYs

ICER vs. BV

($/QALY)

BV

389,802

Reference

7.1469

Reference

Reference

Pembrolizumab

341,865

–47,937

7.9905

0.8437

Dominant

BV = brentuximab vedotin; ICER = incremental cost-effectiveness ratio; QALY = quality-adjusted life-year; vs. = versus.

Source: Sponsor’s pharmacoeconomic submission.1

Sensitivity and Scenario Analysis Results

The sponsor conducted a number of sensitivity and scenario analyses. Key scenario analyses included modelling specific adult and pediatric subpopulations that were ASCT eligible and ASCT ineligible. Furthermore, in the scenario analysis for the ASCT-ineligible subpopulations, both BV and chemotherapy were chosen as pairwise comparators to pembrolizumab. Several of these scenario analyses did not result in pembrolizumab being dominant. In both the adult and pediatric population for ASCT-ineligible subpopulations, the ICER for pembrolizumab versus chemotherapy was $57,508 per QALY gained and $53,014 per QALY gained, respectively, while the ICER for an adult subpopulation who were ASCT eligible was $19,951 per QALY gained.

CADTH Appraisal of the Sponsor’s Economic Evaluation

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

Additional limitations were identified but were not considered to be key limitations.

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

Table 5: Key Assumptions of the Submitted Economic Evaluation (Not Noted as Limitations to the Submission)

Sponsor’s key assumption

CADTH comment

The OS of pembrolizumab is equal to BV

Uncertain pending mature survival data from KEYNOTE-204. |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| ||||||||||||||||||||||||||||||||||||||||

The OS of BV in an adult population is representative of a pediatric population treated with BV or pembrolizumab

Uncertain. |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| |||||||||||||||||| Further, clinical experts consulted by CADTH indicated OS in a pediatric population compared to an adult population would be similar or higher.

PFS for an adult population in KEYNOTE-204 approximates PFS for a pediatric population

Reasonable. The clinical experts consulted by CADTH indicated the PFS results in adults could plausibly be assumed to approximate those of pediatric populations.

BV = brentuximab vedotin; OS = overall survival; PFS = progression-free survival.

CADTH Reanalyses of the Economic Evaluation

Base-Case Results

Several limitations of the sponsor’s submission could not be adequately addressed, resulting in CADTH not being able to conduct a base case for ASCT-ineligible patients treated with pembrolizumab compared to chemotherapy. Therefore, the cost-effectiveness of pembrolizumab in ASCT-ineligible patients could not be determined and is unknown. CADTH was able to conduct a base case for adult and pediatric patients who were ASCT eligible and treated with pembrolizumab compared to BV.

To address limitations identified within the economic model, the CADTH base case was derived by making changes in model parameter values and assumptions, in consultation with clinical experts. Table 6 details each change made to derive the CADTH reanalysis, which was conducted in a step-wise approach to the sponsor’s base case to highlight the impact of each change. For treatment waning, alternate parametric distributions were fitted to PFS, as consulted clinical experts estimated PFS would be equal between pembrolizumab and BV after 10 years (Appendix 4, Figure 4 and Figure 5). For changes in utilities values, CADTH derived utilities from the same source as the sponsor’s base-case values, except the utilities were specific to the pooled analysis instead of being treatment-specific.13

Table 6: CADTH Revisions to the Submitted Economic Evaluation

Stepped analysis

Sponsor’s value or assumption

CADTH value or assumption

Corrections to sponsor’s base case

Dose intensity

0.98

1.0

Errors relating to differential sampling of overall survival for pembrolizumab and BV were corrected

Changes to derive the CADTH base case

1. Population

Full ITT population

ASCT-eligible subpopulation

2. PFS data source

PFS data did not censor those who subsequently received an SCT following treatment. The cost of an SCT was included, based on subgroup specific percentages who received autologous or allogenic SCT.

PFS censored those who subsequently received an SCT following treatment. SCT costs were not included.

3. Treatment waning

Long-term PFS benefits for pembrolizumab over BV continue after the trial duration.

At 10 years, PFS is equal between pembrolizumab and BV.

4. Utilities

Progression-free health state:

  • Pembrolizumab: mean = 0.883 (SE = 0.006)

  • BV: mean = 0.822 (SE = 0.008)

Progressed disease health state:

  • Pembrolizumab: mean = 0.861 (SE = 0.010)

  • BV: mean = 0.766 (SE = 0.015)

Progression-free health state:

  • Mean = 0.857 (SE = 0.005):

Progressed disease health state:

  • Mean = 0.820 (SE = 0.009)

5. Drug wastage

Assumes no drug wastage

Assumes 5% drug wastage

6. Gemcitabine price

1,000 mg vial = $30.0000

2,000 mg vial = $60.0000

1,000 mg vial = $270.0000

2,000 mg vial = $540.0000

7. Subsequent therapy breakdown

Those who start on pembrolizumab subsequent therapy breakdown:

BV (52.1%); pembrolizumab (0.0%); nivolumab (0.0%); gemcitabine (37.9%); none (10.0%)

Those who start on BV subsequent therapy breakdown:

BV (2.9%); pembrolizumab (43.5%); nivolumab (43.5%); gemcitabine (0.0%); none (10.0%)

Those who start on pembrolizumab subsequent therapy breakdown:

BV (35.6%); pembrolizumab (1.9%); nivolumab (4.8%); gemcitabine (47.7%); none (10.0%)

Those who start on BV subsequent therapy breakdown:

BV (4.7%); pembrolizumab (18.2%); nivolumab (20.2%); gemcitabine (46.7%); none (10.0%)

CADTH base case

Reanalysis 1 + 2 + 3 + 4 + 5 + 6

CADTH scenario analysis

Reanalysis 1 + 2 + 3 + 4 + 5 + 6 + 7

ASCT = autologous stem cell transplant; BV = brentuximab vedotin; ITT = intention to treat; PFS = progression-free survival; SCT = stem cell transplant; SE = standard error.

The summary results of the CADTH reanalyses for the adult and pediatric populations are presented in Table 7 and 8, respectively. CADTH undertook a stepped analysis, incorporating each change proposed in Table 6 to the sponsor’s base case to highlight the impact of each change.

In CADTH’s base case for an adult population, pembrolizumab had higher mean costs (incremental: $16,863) and QALYs gained (incremental: 0.0230) than BV. The ICER for pembrolizumab versus BV was $733,624 per QALY gained, and pembrolizumab had a 27.1% probability of being cost-effective at a WTP threshold of $50,000 per QALY gained. A detailed breakdown of the disaggregate results is available in Appendix 4, Table 15. Approximately 20% of the total QALYs gained in the model were generated within the trial period (1.53 QALYs), while the remaining 80% were generated over the extrapolated post-trial period (5.91 QALYs).

Table 7: Summary of the Stepped Analysis of the CADTH Adult Reanalysis Results

Stepped analysis

Drug

Total costs ($)

Total QALYs

ICER ($/QALYs)

Sponsor’s corrected base case

BV

426,419

6.9820

Reference

Pembrolizumab

399,090

7.8333

Dominant

CADTH reanalysis 1 ASCT eligible

BV

412,496

7.0806

Reference

Pembrolizumab

427,059

7.8571

18,754

CADTH reanalysis 2 PFS data source

BV

411,353

6.9834

Reference

Pembrolizumab

394,797

7.8031

Dominant

CADTH reanalysis 3 Treatment waning

BV

427,208

7.0723

Reference

Pembrolizumab

405,041

7.8837

Dominant

CADTH reanalysis 4 Disease-specific utilities

BV

426,410

7.4626

Reference

Pembrolizumab

399,379

7.5388

Dominant

CADTH reanalysis 5 Drug wastage

BV

445,113

7.0104

Reference

Pembrolizumab

412,631

7.8556

Dominant

CADTH reanalysis 6 Gemcitabine cost

BV

426,695

7.0337

Reference

Pembrolizumab

400,559

7.8446

Dominant

CADTH reanalysis 7 Subsequent therapy breakdown

BV

378,411

7.0089

Reference

Pembrolizumab

393,123

7.8046

18,489

CADTH base case (1 + 2 + 3 + 4 + 5 + 6)

BV

421,300

7.4370

Reference

Pembrolizumab

438,163

7.4600

733,624

CADTH scenario analysis (1 + 2 + 3 + 4 + 5 + 6 + 7)

BV

375,409

7.4678

Reference

Pembrolizumab

428,777

7.4936

2,071,825

ASCT = autologous stem cell transplant; BV = brentuximab vedotin; ICER = incremental cost-effectiveness ratio; PFS = progression-free survival; QALY = quality-adjusted life-year.

Note: The reanalysis is based on the publicly available prices of the comparator treatments.

In CADTH’s base case for the pediatric population, pembrolizumab had lower mean costs (incremental: –$16,685) and higher QALYs gained (incremental: 0.0393) than BV. The ICER indicated pembrolizumab was dominant (i.e., cost savings and more QALYs gained) versus BV, and pembrolizumab had a 46.7% probability of being cost-effective at a WTP threshold of $50,000 per QALY gained. A detailed breakdown of the disaggregate results is available in Appendix 4, Table 14. Similar to the adult population, 20% of the total QALYs gained in the model were generated within the trial period (1.53 QALYs), while the remaining 80% were generated over the extrapolated post-trial period (5.99 QALYs). This analysis is reliant on assuming that data from the adult population can be used for pediatrics.

Table 8: Summary of the Stepped Analysis of the CADTH Pediatric Reanalysis Results

Stepped analysis

Drug

Total costs ($)

Total QALYs

ICER ($/QALYs)

Sponsor’s corrected base case

BV

394,426

7.0693

Reference

Pembrolizumab

344,384

7.8933

Dominant

CADTH reanalysis 1 ASCT eligible

BV

368,167

7.0300

Reference

Pembrolizumab

322,413

7.8567

Dominant

CADTH reanalysis 2 PFS data source

BV

354,795

7.0444

Reference

Pembrolizumab

319,645

7.8924

Dominant

CADTH reanalysis 3 Treatment waning

BV

393,995

7.0647

Reference

Pembrolizumab

349,710

7.8666

Dominant

CADTH reanalysis 4 Disease-specific utilities

BV

393,800

7.4795

Reference

Pembrolizumab

344,619

7.5664

Dominant

CADTH reanalysis 5 Drug wastage

BV

453,580

7.0716

Reference

Pembrolizumab

428,608

7.8797

Dominant

CADTH reanalysis 6 Gemcitabine cost

BV

392,692

7.0284

Reference

Pembrolizumab

344,168

7.8387

Dominant

CADTH reanalysis 7 Subsequent therapy breakdown

BV

363,453

7,0771

Reference

Pembrolizumab

340,893

7,9054

Dominant

CADTH base case (1 + 2 + 3 + 4 + 5 + 6)

BV

420,896

7.4861

Reference

Pembrolizumab

404,211

7.5254

Dominant

CADTH scenario analysis (1 + 2 + 3 + 4 + 5 + 6 + 7)

BV

374,956

7.4783

Reference

Pembrolizumab

395,735

7.5174

532,115

ASCT = autologous stem cell transplant; BV = brentuximab vedotin; ICER = incremental cost-effectiveness ratio; PFS = progression-free survival; QALY = quality-adjusted life-year.

Note: The reanalysis is based on the publicly available prices of the comparator treatments.

Scenario Analysis Results

CADTH conducted a scenario analysis with alternate subsequent therapy breakdowns for those patients who progress. In this analysis pembrolizumab was less cost-effective with an ICER of $2,071,825 relative to BV in the adult population and $532,115 in the pediatric population.

A price reduction analysis was performed based on the sponsor’s and CADTH’s reanalysis. Based on the CADTH base case, a price reduction of approximately 13% is required to make pembrolizumab cost-effective at a WTP threshold of $50,000 per QALY gained in an adult ASCT-eligible population (Table 9). Despite the high ICER, there is a relatively small price reduction as pembrolizumab is achieves cost savings across various cost categories (e.g., progressed disease, subsequent treatment, and adverse events) with the exception of drug acquisition costs. Furthermore, small incremental QALY gains of 0.023 result in changes in incremental costs, drastically altering the ICER. A price reduction was conducted in the pediatric population given the ICER was dominant (cost savings and more QALYs gained).

A price reduction analysis was performed based on CADTH’s scenario analysis that used a different breakdown of subsequent therapy. In this analysis, the cost savings from subsequent therapy use are substantially lower. Therefore, a higher price reduction of approximately 29% is required to ensure cost-effectiveness at a $50,000 per QALY threshold.

Table 9: CADTH Price Reduction Analysis for Adults (Deterministic)

Price reduction

ICERs for pembrolizumab vs. BV ($/QALY)

ADTH adult reanalysis (subsequent therapy use derived from trial)

Sponsor base case

CADTH adult reanalysis

No price reduction

Dominant

733,624

2,071,825

10%

Dominant

270,319

1,649,910

13%

Dominant

54,300

1,393,631

14%

Dominant

Dominant

1,308,205

20%

Dominant

Dominant

795,648

28%

Dominant

Dominant

112,239

29%

Dominant

Dominant

26,813

30%

Dominant

Dominant

Dominant

BV = brentuximab vedotin; ICER = incremental cost-effectiveness ratio; QALY = quality-adjusted life-year; vs. = versus.

Issues for Consideration

Overall Conclusions

Evidence suggests pembrolizumab provides statistically and clinically significant improvements in PFS |||||||||||||||||||||||||||||||||||| ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||. Across all studies, BV patients were generally less likely to experience adverse events but more likely to discontinue therapy due to an adverse event. No comparators beyond BV were evaluated. Therefore, the comparative evidence of pembrolizumab to chemotherapy in an ASCT-ineligible subpopulation is unknown, due to a lack of direct or indirect evidence for PFS and OS.

CADTH addressed several key limitations of the sponsor’s model by selecting appropriate comparators for the Canadian context; evaluating an ASCT-eligible subpopulation with comparative evidence; using PFS data that censored upon SCT events; considering a treatment effect waning; changing treatment-specific utilities to disease-specific; and incorporating drug wastage. According to CADTH’s reanalyses, in adult patients, pembrolizumab is associated with an ICER of $733,624 per QALY gained when compared to BV, and in a pediatric population pembrolizumab is dominant (i.e., cost saving and associated with more QALYs gained). In adults, a price reduction of approximately 13% is required for pembrolizumab to be cost-effective at a WTP threshold of $50,000 per QALY. This result is contingent on pembrolizumab generating approximately $43,000 of cost savings due to patients who receive pembrolizumab first line and not receiving immunotherapies such as nivolumab and pembrolizumab second line. There is a significant amount of outstanding uncertainty regarding the degree of these cost savings. A scenario analysis that more closely matched subsequent therapy data from the KEYNOTE-204 trial found the ICER in adults increased to $2,071,825 per QALY. At this level, a 29% price reduction is needed to achieve a $50,000 per QALY threshold.

The probability that pembrolizumab is cost-effective at a WTP threshold of $50,000 per QALY was low in both the adult (27.1%) and pediatric (46.7%) populations. The low probability of pembrolizumab being cost-effective in a pediatric population differs from the mean ICER, which indicated pembrolizumab was dominant (less costly and more effective) versus BV. This finding highlights the uncertainty across probabilistic iterations, and the extreme ICERs generated due to marginal differences in costs and QALYs between pembrolizumab and BV.

The conclusions of CADTH’s analysis are specific to an ASCT-eligible population. CADTH was unable to assess the cost-effectiveness of pembrolizumab in an ASCT-ineligible population due to an absence of comparative evidence between pembrolizumab and chemotherapy. In Canada, individuals who are ineligible for ASCT rarely receive BV, therefore chemotherapy is the most relevant comparator. Drug costs for pembrolizumab are substantially higher relative to chemotherapy, as such information on this comparison is needed to assess the value of pembrolizumab in this subgroup. Finally, results for the pediatric population are almost entirely extrapolated from the adult data as well, so any uncertainty with that extrapolation will impact the robustness of the pediatric results.

References

1.Pharmacoeconomic evaluation [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: keytruda (pembrolizumab), 200 mg intravenously (IV) Q3W (for adult patients); 2 mg/kg IV Q3W, up to a maximum of 200 mg (for pediatric patients). Kirkland (QC): Merck Canada Inc.; 2021 Jan 29.

2.Keytruda (pembrolizumab): powder for solution for infusion 50 mg, solution for infusion 100 mg/4 mL vial; Antineoplastic agent, monoclonal antibody [product monograph]. Kirkland (QC): Merck Canada Inc.; 2021 Jun 4.

3.Kuruvilla J, Ramchandren R, Santoro A, et al. Pembrolizumab versus brentuximab vedotin in relapsed or refractory classical Hodgkin lymphoma (KEYNOTE-204): an interim analysis of a multicentre, randomised, open-label, phase 3 study. Lancet Oncol. 2021;22(4):512-524. PubMed

4.Geoerger B, Kang HJ, Yalon-Oren M, et al. Pembrolizumab in paediatric patients with advanced melanoma or a PD-L1-positive, advanced, relapsed, or refractory solid tumour or lymphoma (KEYNOTE-051): interim analysis of an open-label, single-arm, phase 1-2 trial. Lancet Oncol. 2020;21(1):121-133. PubMed

5.Chen R, Gopal AK, Smith SE, et al. Five-year survival and durability results of brentuximab vedotin in patients with relapsed or refractory Hodgkin lymphoma. Blood. 2016;128(12):1562-1566. PubMed

6.CADTH pCODR Expert Review Committee final economic guidance report: pembrolizumab (Keytruda) for renal cell carcinoma. 2020 Apr 2. https://cadth.ca/sites/default/files/pcodr/Reviews2020/10185PembrolizumabRCC_fnEGR_NOREDACT-ABBREV_Post02Apr2020_final.pdf. Accessed May 17, 2021.

7.CADTH Drug Reimbursement Review pharmacoeconomic report: brentuximab vedotin (Adcetris) for the treatment of previously untreated patients with stage IV Hodgkin lymphoma, in combination with doxorubicin, vinblastine, and dacarbazine. Ottawa (ON): CADTH; 2020 Dec 3: https://www.cadth.ca/sites/default/files/pcodr/Reviews2020/10214BrentuximabVedotinHLAVD_fnEGR_NOREDACT-ABBREV_03Dec2020_final.pdf. Accessed 2021 May 11.

8.Hirt C, Iannazzo S, Chiroli S, et al. Cost effectiveness of the third-generation tyrosine kinase inhibitor (TKI) ponatinib, vs. second-generation TKIs or stem cell transplant, as third-line treatment for chronic-phase chronic myeloid leukemia. Appl Health Econ Health Policy. 2019;17(4):555-567. PubMed

9.Drug Reimbursement Review sponsor submission: Keytruda (pembrolizumab), 200 mg intravenously (IV) Q3W (for adult patients); 2 mg/kg IV Q3W, up to a maximum of 200 mg (for pediatric patients) [internal sponsor's package]. Kirkland (QC): Merck Canada Inc.; 2021 Jan 29.

10.CADTH pCODR Expert Review Committee final recomendation: brentuximab vedotin (Adcetris). 2019 Mar 7. https://cadth.ca/sites/default/files/pcodr/Reviews2019/10145BrentuximabHL_FnRec_approvedbyChair_Post_07Mar2019_final.pdf. Accessed 2021 May 7.

11.Guidelines for the economic evaluation of health technologies: Canada. CADTH methods and guidelines. 4th ed. Ottawa (ON): CADTH; 2017: https://www.cadth.ca/sites/default/files/pdf/guidelines_for_the_economic_evaluation_of_health_technologies_canada_4th_ed.pdf. Accessed 2021 May 11.

12.Gridelli C, Gallo C, Shepherd FA, et al. Gemcitabine plus vinorelbine compared with cisplatin plus vinorelbine or cisplatin plus gemcitabine for advanced non-small-cell lung cancer: a phase III trial of the Italian GEMVIN Investigators and the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2003;21(16):3025-3034. PubMed

13.Clinical study report: P204V01MK3475. A phase III, randomized, open-label, clinical trial to compare pembrolizumab with brentuximab vedotin in subjects with relapsed or refractory classical Hodgkin lymphoma. [internal sponsor's report]. Kenilworth (NJ): Merck Sharp & Dohme Corp; 2020 Apr 10.

14.DeltaPA. [Ottawa (ON)]: IQVIA; 2021: https://www.iqvia.com/. Accessed 2021 May 11.

15.Exceptional Access Program (EAP). Toronto (ON): Ontario Ministry of Health; Ontario Ministry of Long-Term Care; 2020: https://www.health.gov.on.ca/en/pro/programs/drugs/odbf/odbf_except_access.aspx. Accessed 2021 May 11.

16.Ontario drug benefit formulary/comparative drug index. Toronto (ON): Ontario Ministry of Health, Ontario Ministry of Long-Term Care; 2020: https://www.formulary.health.gov.on.ca/formulary/. Accessed 2021 May 11.

17.Budget impact analysis [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: keytruda (pembrolizumab), 200 mg intravenously (IV) Q3W (for adult patients); 2 mg/kg IV Q3W, up to a maximum of 200 mg (for pediatric patients). Kirkland (QC): Merck Canada Inc.; 2021 Jan 29.

Appendix 1: Cost Comparison Table

Note that this appendix has not been copy-edited.

The comparators presented in the Table 10 have been deemed to be appropriate based on feedback from clinical experts. Comparators may be recommended (appropriate) practice or actual practice. Existing Product Listing Agreements are not reflected in the table and as such, the table may not represent the actual costs to public drug plans.

Table 10: CADTH Cost Comparison Table for Relapsed or Refractory Classical Hodgkin Lymphoma

Treatment

Strength / concentration

Form

Price

Recommended dosage

Daily cost

28-day cost

Pembrolizumab

(Keytruda)a

25 mg/mL

100 mg

Solution for IV infusion

$4,400.0000

Adults: 200mg once every 3 weeks

$419.05

$11,733

Pediatrics: 2mg/kg once every 3 weeks

$419.05

$11,733

Anti-PD-1 monoclonal antibodies

Nivolumab

(Opdivo)

10 mg/mL

40 mg

100 mg

Vial for IV infusion

$782.2200

$1,955.5600

Adults: 6 mg/kg once every 2 weeks

$335.24 to $419.05

$9,387 to $11,733

Pediatrics: 3 mg/kg every 2 weeks

$279.36

$7,822

Antibody drug conjugate

Brentuximab vedotin

(Adcetris)

4.76 mg/mL

50 mg

Powder for IV infusion

$4,840.0000

Adults: 1.8 mg/kg once every 3 weeks

$691.43

$19,360

Pediatrics: 1.8 mg/kg once every 3 weeks

$691.43

$19,360

Nucleoside analogue

Gemcitabine

40 mg/mL

1,000 mg

2,000 mg

Solution for IV infusion

$270.0000

$540.0000

Adults: 1,000 mg/m2 thrice every 4 weeks

$57.86

$1,620

Pediatrics: 1,000 mg/m2 twice every 3 weeks

$51.43

$1,440

Immunomodulatory agent

Lenalidomide

(Revlimid)b

2.5 mg

5 mg

10 mg

15 mg

20 mg

25 mg

Cap

$329.5000

$340.0000

$361.0000

$382.0000

$403.0000

$424.0000

Adults: 10 mg on day 1-21 every 4 weeks

$270.75

$7,581

Alkylating agents

Bendamustine

(Treanda)

5 mg/mL

25 mg

100 mg

Powder for IV infusion

$265.6300

$1,062.5000

Adults: 120 mg/m2 twice every 3 weeks

$252.98 to $303.57

$7,083 to $8,500

Pediatrics: 120 mg/m2 twice every 3 weeks

$202.38

$5,667

Brentuximab + gemcitabine

Brentuximab vedotin

(Adcetris)

4.76 mg/mL

50 mg

Powder for IV infusion

$4,840.0000

Pediatric: 1.8 mg/kg once every 3 weeks

$691.43

$19,360

Gemcitabine

40 mg/mL

1,000 mg

2,000 mg

Solution for IV infusion

$270.0000

$540.0000

Pediatric: 1,000 mg/m2 twice every 3 weeks

$51.43

$1,440

Brentuximab + gemcitabine regiment cost per 28 days

$742.86

$20,800

Brentuximab + bendamustine

Brentuximab vedotin

(Adcetris)

4.76 mg/mL

50 mg

Powder for IV infusion

$4,840.0000

Adults: 1.8 mg/kg once every 3 weeks

$691.43

$19,360

Bendamustine

(Treanda)

5 mg/mL

25 mg

100 mg

Powder for IV infusion

$265.6300

$1,062.5000

Adults: 90 mg/m2 twice every 3 weeks

$202.38

$5,667

Brentuximab + bendamustine regiment cost per 28 days

$893.81

$25,027

GV

Gemcitabine

40 mg/mL

1,000 mg

2,000 mg

Solution for IV infusion

$270.0000

$540.0000

Pediatric: 1,000 mg/m2 twice every 3 weeks

$51.43

$1,440

Vinorelbine

10 mg/mL

10 mg

50 mg

Solution for IV infusion

$80.0000

$400.0000

Pediatric: 25 mg/m2 twice every 3 weeks

$30.48

$853

GV regiment cost per 28 days

$81.90

$2,293

IGEV

Ifosfamide

50 mg/mL

1,000 mg

3,000 mg

Vial for IV infusion

$129.1700

$395.5599

Adults: 2000 mg/m2 4 times every 3 weeks

$150.69

$4,219

Mesna

100 mg/mL

1,000 mg

Solution for IV infusion

$227.8900

Adults: 2000 mg/m2 4 times every 3 weeks

$164.95

$4,619

Gemcitabine

40 mg/mL

1,000 mg

2,000 mg

Solution for IV infusion

$270.0000

$540.0000

Adults: 800 mg/m2 twice every 3 weeks

$51.43

$1,440

Vinorelbine

10 mg/mL

10 mg

50 mg

Solution for IV infusion

$80.0000

$400.0000

Adults: 20 mg/m2 once every 3 weeks

$15.24

$427

IGEV regiment cost per 28 days

$382.31

$10,705

GDP

Gemcitabine

40 mg/mL

1,000 mg

2,000 mg

Solution for IV infusion

$270.0000

$540.0000

Adults: 1,000 mg/m2 twice every 3 weeks

$51.43

$1,440

Dexamethasonec

0.5 mg

4 mg

Tab

$0.1564

$0.3046

Adults: 40 mg 4 times every 3 weeks

$0.58

$16

Cisplatin

1 mg/mL

50 mg

100 mg

Vial for IV infusion

$135.0000

$270.0000

Adults: 75 mg/m2 once every 3 weeks

$36.64

$1,026

GDP regiment cost per 28 days

$88.65

$2,482

ICE

Ifosfamide

50 mg/mL

1,000 mg

3,000 mg

Vial for IV infusion

$129.1700

$395.5599

Adults: 5,000 mg/m2 once every 3 weeks

$75.34

$2,110

Mesna

100 mg/mL

1,000 mg

Solution for IV infusion

$227.8900

Adults: 5,000 mg/m2 once every 3 weeks

$103.09

$2,887

Carboplatin

10 mg/mL

50 mg

150 mg

450 mg

600 mg

Solution for IV infusion

$70.0000

$210.0000

$599.9985

$775.0020

Adults: 400 mg/m2 once every 3 weeks

$57.14

$1,600

Etoposide

50 mg

Cap

$ 41.5875

Adults: 100 mg thrice every 3 weeks

$11.88

$333

ICE regiment cost per 28 days

$247.46

$6,929

DHAP

Dexamethasonec

0.5 mg

4 mg

Tab

$0.1564

$0.3046

Adults: 40 mg 4 times every 3 weeks

$0.58

$16

Cytarabine

100 mg/mL

500 mg

2,000 mg

Solution for IV infusion

$76.8500

$306.5000

Adults: 2000 mg/m2 twice every 3 weeks

$55.46

$1,553

Cisplatin

1 mg/mL

50 mg

100 mg

Vial for IV infusion

$135.0000

$270.0000

Adults: 100 mg/m2 once every 3 weeks

$48.86

$1,368

DHAP regiment cost per 28 days

$104.90

$2,937

IV = intravenous; NA = not applicable.

Note: All prices are IQVIA Delta PA wholesale list prices (accessed May 2021),14 unless otherwise indicated, and do not include dispensing fees or markups. Costs assume a body weight of 76.45 kg and 58.5 kg and body surface area of 1.9 m2 and 1.6 m2 for the adult and pediatric populations, respectively. Costs include wastage of unused medication in vials when indicated in the product monograph as single-use.

aSponsor’s submitted price.1

bOntario Exceptional Access Program e-formulary (accessed May 2021).15

cOntario Drug Benefit e-formulary (accessed May 2021).16

Appendix 2: Submission Quality

Note that this appendix has not been copy-edited.

Table 11: Submission Quality

Description

Yes/No

Comments

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

No

Given differing interventions across subpopulations, the base-case population is not relevant (i.e., BV is not a relevant comparator in those ASCT ineligible). Chemotherapy is the relevant comparator for the ASCT-ineligible subpopulation, but there was a lack of direct or indirect evidence to adequately assess cost-effectiveness.

Model has been adequately programmed and has sufficient face validity

No

The model’s programming has multiple limitations. The time horizon can only be altered by 5-year increments from 15 years to 60 years. Treatment waning has arbitrary limits to the start data (5 years) and total duration (12 years). Finally, utility data are treatment-specific not disease-specific, which does not adhere to CADTH best practices which recommends utilities reflect the health states within the model and not be specific to treatments.

Model structure is adequate for decision problem

Yes

Model structure follows standard partition survival models.

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

Yes

None

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

Yes

None

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

No

The model was unnecessarily complex, and information in the report was difficult to locate. Specifically, there were cases it referred to nonexistent sections of the report for further clarification on the methods.

Appendix 3: Additional Information on the Submitted Economic Evaluation

Note that this appendix has not been copy-edited.

Figure 1: Model Structure

A diagrammatic representation of a partition survival model that details how patients move through the model states.

OS = overall survival; PD = progressed disease; PF = progression-free; PFS = progression-free survival.

Source: Figure 3 in the sponsor’s pharmacoeconomic report (p. 37).1

Figure 2: PFS Extrapolations for Pembrolizumab Versus BV (52-Week Cut-Off Point)

Graph presenting the proportion of patients who are progression free at various time points within the model, dependent on whether they receive BV or pembrolizumab.

PFS = Progression-free survival; BV = brentuximab vedotin.

Table 12: Disaggregated Results of Adult Population With ITT Base Case

Parameter

Pembrolizumab

BV

Differential values

Discounted life-years

Total LYs

8.9927

8.9873

0.0054

By health state

   PF

4.8482

2.4885

2.3597

   PD

4.1445

6.4988

–2.3543

Discounted QALYs

Total QALYs

7.8508

7.0183

0.8325

By health state

   PF

4.2827

2.0462

2.2365

   PD

3.5681

4.9721

–1.4040

Age-related decrement

0.0000

0.0000

0.0000

AEs

0.0000

0.0000

0.0000

Incremental QALYs generated within trial period

1.5949

1.4482

0.1467

Incremental QALYs generated after trial period

6.2559

5.5701

0.6858

Discounted costs

Total costs

396,223

424,313

–28,090

PF costs ($)

54,743

28,099

26,644

PD costs ($)

96,545

151,387

–54,842

Terminal costs ($)

20,190

20,195

–5

Acquisition costs ($)

159,126

106,487

52,639

Administration costs ($)

1,437

675

763

Subsequent treatment cost ($)

37,625

90,231

–52,606

AE costs ($)

1,900

2,328

–427

SCT costs ($)

24,656

24,912

–256

Incremental cost per QALY gain

Dominant

Table 13: Disaggregated Results of Pediatric Population With ITT Base Case

Parameter

Pembrolizumab

BV

Differential values

Discounted life-years

Total LYs

9.0362

9.0077

0.0284

By health state

   PF

4.8182

2.5018

2.3164

   PD

4.2180

6.5059

–2.2880

Discounted QALYs

Total QALYs

7.8808

7.0417

0.8391

By health state

   PF

4.2486

2.0566

2.1921

   PD

3.6322

4.9851

–1.3529

Age-related decrement

0.0000

0.0000

0.0000

AEs

0.0000

0.0000

0.0000

Incremental QALYs generated within trial period

1.5934

1.4478

0.1456

Incremental QALYs generated after trial period

6.2875

5.5939

0.6936

Discounted costs

Total costs

343,540

391,682

–48,142

PF costs ($)

54,405

28,249

26,156

PD costs ($)

98,256

151,553

–53,298

Terminal costs ($)

20,000

20,030

-30

Acquisition costs ($)

93,166

81,604

11,562

Administration costs ($)

1,436

675

761

Subsequent treatment cost ($)

28,945

59,634

–30,689

AE costs ($)

1,901

2,335

–435

SCT costs ($)

45,433

47,603

–2,170

Incremental cost per QALY gain

Dominant

Figure 3: Errors in Treatment Waning for PFS Extrapolations for Pembrolizumab Versus BV (52-Week Cut-Off Point)

Graph presenting the proportion of patients who are progression free over time based on the sponsor’s model. The graph shows an implausible scenario where patients who take pembrolizumab start to move from progressed to progression free after 20 years.

PFS = progression-free survival; BV = brentuximab vedotin.

Note: PFS was modelled using the secondary dataset that censors patients upon SCT event, and pembrolizumab was given a Weibull distribution.

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

Note that this appendix has not been copy-edited.

Figure 4: PFS Extrapolations — Adult ASCT-Eligible Population

Graph presenting the proportion of adult patients who are progression free at various time points within the model, dependent on whether they receive BV or pembrolizumab.

Figure 5: PFS Extrapolations — Pediatric ASCT-Eligible Population

Graph presenting the proportion of pediatric patients who are progression free at various time points within the model, dependent on whether they receive BV or pembrolizumab.

PFS = progression-free survival; BV = brentuximab vedotin.

Table 14: Disaggregated Summary of CADTH’s Economic Evaluation Results — Pediatric ASCT-Eligible Population

Parameter

Pembrolizumab

BV

Incremental

Discounted LYs

Total

9.0223

9.0223

0.0000

By health state or data source

   Progression free

3.4194

2.3159

1.1035

   Progressed disease

5.6029

6.7065

–1.1035

Discounted QALYs

Total

7.5254

7.4861

0.0393

By health state or data source

   Progression free

2.9269

1.9834

0.9434

   Progressed disease

4.5985

5.5027

–0.9041

Discounted costs ($)

Total

404,211

420,896

–16,685

   Progression free

38,610

26,150

12,461

   Progressed disease

130,518

156,224

–25,706

   Terminal

20,014

20,014

0

   Acquisition

166,741

123,414

43,326

   Administration

1,424

737

687

   Subsequent treatment

45,000

92,019

–47,020

   Adverse events

1,904

2,337

–433

ICER ($/QALY)

Dominant (less costly and more effective)

ICER = incremental cost-effectiveness ratio; LY= life-year; QALY = quality-adjusted life-year; BV = brentuximab vedotin.

Table 15: Disaggregated Summary of CADTH’s Economic Evaluation Results — Adult ASCT-Eligible Population

Parameter

Pembrolizumab

BV

Incremental

Discounted LYs

Total

8.9534

8.9534

0.0000

By health state or data source

   Progression free

3.6542

3.0666

0.5876

   Progressed disease

5.2992

5.8868

–0.5876

Discounted QALYs

Total

7.4600

7.4370

0.0230

By health state or data source

   Progression free

3.1279

2.6229

0.5050

   Progressed disease

4.3321

4.8141

–0.4820

Discounted costs ($)

Total

438,163

421,300

16,863

   Progression free

41,262

34,627

6,635

   Progressed disease

123,442

137,131

–13,689

   Terminal

20,230

20,230

0

   Acquisition

201,918

135,581

66,338

   Administration

1,706

737

968

   Subsequent treatment

47,718

90,661

–42,943

   Adverse events

1,887

2,334

–447

ICER ($/QALY)

$733,624

ICER = incremental cost-effectiveness ratio; LY= life-year; QALY = quality-adjusted life-year; BV = brentuximab vedotin.

Appendix 5: Submitted BIA and CADTH Appraisal

Note that this appendix has not been copy-edited.

Table 16: Summary of Key Take-Aways

Key Take-Aways of the BIA

  • CADTH identified the following key limitations with the sponsor’s analysis:

    • The market share of BV in an ASCT-ineligible population is uncertain.

    • The current utilization of clinical trials is uncertain, as is future use of clinical trials pending a new treatments entrance to the market; therefore, clinical trial use was excluded.

    • Pembrolizumab’s uptake rate for its first year was low.

    • Pembrolizumab is indicated as a single-use drug, therefore drug wastage was incorporated.

    • The price of gemcitabine was too low and unrepresentative of the Canadian list price.

  • CADTH’s reanalysis removed clinical trials’ market share and redistributed it among existing treatments, increased the first-year uptake rate of pembrolizumab, included drug wastage, and revised the price of gemcitabine to the Canadian list price. BV market shares were changed as a scenario analysis. Based on the CADTH base case, the expected budget impact for funding pembrolizumab is $305,213 in year 1, $2,070,116 in year 2, and $3,035,408 in year 3, for a total 3-year budget impact of $5,410,737.

Summary of Sponsor’s BIA

The submitted budget impact analysis (BIA) assessed the introduction of pembrolizumab, as monotherapy, for adults and pediatric patients with relapsed/refractory classical Hodgkin lymphoma (cHL), for third-line treatment in those who relapsed post-ASCT or as second-line treatment in those ineligible for ASCT. The analysis was taken from the perspective of the Canadian public drug plans using an epidemiologic-based approach, with only drug acquisition costs considered. A 3-year time horizon was used, from 2022 to 2024, with 2021 as a base year.

A summary of the sponsor’s derivation of the eligible population size is presented in Figure 6, and key inputs to the BIA are documented in Table 17. The sponsor estimated the current population using an epidemiologic approach, derived from non-Canadian and Canadian publications, health technology assessment recommendations, and clinical expert opinions. The incidence of Hodgkin lymphoma patients in 2022 was estimated at 761 patients, of which 95% would have the classical subtype. Clinician input indicated that all cHL patients receive first-line ABVD (Adriamycin-bleomycin-vinblastine-dacarbazine) treatment because of its high cure rate. Of that treated population, Canadian data indicated the 5-year risk of relapse was 18.1%, which was used to calculate the relapse/refractory cHL population. Based on that same data in relapse/refractory cHL patients, 78% would undergo ASCT while the remaining was assumed to represent the ASCT-ineligible subpopulation. Each subpopulation was further stratified into adult and pediatric patients based on Statistics Canada data on Hodgkin lymphoma (to allow for differential drug dosing and costing), where approximately 12% of all patients were considered pediatric.

The sponsor’s submission had a reference scenario in which patients were initially treated with either BV or chemotherapy, and a new drug scenario in which pembrolizumab was reimbursed. The sponsor also conducted various sensitivity analyses where wastage, subsequent SCT costs, and administration costs were included.

Figure 6: Sponsor’s Estimation of the Size of the Eligible Population

A flowchart showing how the population size eligible for treatment with pembrolizumab in the submitted indication was derived.

Source: Sponsor’s budget impact submission – Figure 2.17

Table 17: Summary of Key Model Parameters

Parameter

Sponsor’s estimate

Target population

Incidence of Hodgkin lymphoma

Relapse/refractory after first-line therapy

ASCT-eligible patients

Yearly growth

761

18%

78%

0%

Number of patients eligible for drug under review

ASCT eligible (years 1 / 2 / 3)

ASCT ineligible (years 1 / 2 / 3)

26 / 26 / 26

26 / 26 / 26

Market uptake (3 years)

Uptake (reference scenario in years 1 / 2 / 3)

ASCT eligible:

Pembrolizumab

BV

Chemotherapy

Clinical trial

0% / 0% / 0%

83% / 83% / 83%

5% / 5% / 5%

12% / 12% / 12%

ASCT ineligible

Pembrolizumab

BV

Chemotherapy

Clinical trial

2% / 2% / 2%

32% / 32% / 32%

63% / 62% / 62%

3% / 3% / 3%

Uptake (new drug scenario in years 1 / 2 / 3)

ASCT eligible:

Pembrolizumab

BV

Chemotherapy

Clinical trial

45% / 86% / 86%

45% / 12% / 12%

3% / 1% / 1%

7% / 2% / 2%

ASCT ineligible:

Pembrolizumab

BV

Chemotherapy

Clinical trial

47% / 86% / 86%

17% / 4% / 4%

34% / 9% / 9%

2% / 0% / 0%

Cost of treatment (per patient)

Cost of treatment over cycle

Pembrolizumab (adult) – Q3W

Pembrolizumab (pediatric) – Q3W

Brentuximab vedotin (hybrid)a – Q3W

Gemcitabine (adult) – (3 x Q4W)

Gemcitabine (pediatric) – (2 x Q3W)

Clinical trial

$8,641.60

$5,055.34

$12,712.66

$167.92

$94.27

$0.00

BV = brentuximab vedotin; ASCT = autologous stem cell transplant; Q3W = every 3 weeks, Q4W = every 4 weeks.

aHybrid: cost was determined by calculating 12% of the average dose using the pediatric body weight or body surface area,

Summary of the Sponsor’s BIA Results

From the perspective of Canadian public drug plans, the estimated budget of reimbursing pembrolizumab for the treatment of relapsed/refractory cHL patients for both third-line treatment in those who relapsed post-ASCT and as second-line treatment in those ineligible for ASCT, is expected to be $277,229 in Year 1, $2,038,724 in Year 2, and $3,538,473 in Year 3, with a 3-year budget impact of $5,854,426. Note, these costs include both the initial and subsequent treatments.

CADTH Appraisal of the Sponsor’s BIA

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

Additional limitations were identified but were not considered to be key limitations. These limitations include:

CADTH Reanalyses of the BIA

Based on the limitations identified, CADTH’s base case included: the removal of clinical trials as an intervention, an increased pembrolizumab uptake rate in year 1, accounting for drug wastage, and correcting gemcitabine’s price to the Canadian list price. A scenario analysis explored the reduction in BV market share (Table 20).

Table 18: CADTH Revisions to the Submitted Budget Impact Analysis

Stepped analysis

Sponsor’s value or assumption

CADTH value or assumption

Corrections to sponsor’s base case

Dose intensity

0.98

1.0

Changes to derive the CADTH base case

1. Clinical trials

ASCT eligible:

BV = 82.7%

Chemotherapy = 5.3%

Clinical trials = 12.1%

ASCT ineligible:

Pembrolizumab = 2.3%

BV = 31.5%

Chemotherapy = 62.7%

Clinical trials = 3.4%

ASCT eligible:

BV = 94%

Chemotherapy = 6.0%

Clinical trials = 0.0%

ASCT ineligible:

Pembrolizumab = 2.4%

BV = 32.6%

Chemotherapy = 65.0%

Clinical trials = 0.0%

2. Pembrolizumab uptake rate

Linear extrapolation

Year 1 ASCT eligible: 45%

Year 1 ASCT ineligible: 47%

Logarithmic extrapolation

Year 1 ASCT eligible: 67%

Year 1 ASCT ineligible: 68%

3. Drug wastage

Assumes no drug wastage

Assumes 5% drug wastage

4. Gemcitabine price

Cost per mg = $0.03

Cost per mg = $0.27

5. BV market share

32%

15%

CADTH base case

Reanalysis 1 + 2 + 3 + 4

CADTH scenario analysis

Reanalysis 1 + 2 + 3 + 4 + 5

ASCT = autogenic stem cell transplant; BV = brentuximab vedotin.

The results of the CADTH step-wise reanalysis are presented in summary format in Table 1 and a more detailed breakdown is presented in Table 2. Based on the CADTH base case, the expected budget impact for funding pembrolizumab is $305,213 in year 1, $2,070,116 in year 2, and $3,035,408 in year 3, for a total 3-year budget impact of $5,410,737. Within the total 3-year budget impact, $1,503,571 was from the third-line ASCT-eligible subpopulation and $3,907,166 was from the second-line ASCT-ineligible subpopulation.

CATH conducted a scenario analysis where the BV market share was reduced to 15%. In this analysis the expected budget impact for funding pembrolizumab is $518,795 in year 1, $2,551,263 in year 2, and $3,587,068 in year 3, for a total 3-year budget impact of $6,657,127.

Table 19: Summary of the CADTH Reanalyses of the BIA

Stepped analysis

Three-year total

Sponsor’s corrected base case

$5,961,737

CADTH reanalysis 1 – remove clinical trials

$5,056,607

CADTH reanalysis 2 – increase pembrolizumab uptake rate

$6,923,637

CADTH reanalysis 3 – include drug wastage

$5,961,270

CADTH reanalysis 4 – revise gemcitabine price

$5,560,984

CADTH reanalysis 5 – reduce BV market share

$7,100,120

CADTH base case (1 + 2 + 3 + 4)

$5,410,737

CADTH scenario analysis (1 + 2 + 3 + 4 + 5)

$6,657,127

BIA = budget impact analysis.

Note: This analysis is based on the publicly available prices of the comparator treatments.

Table 20: Detailed Breakdown of the CADTH Reanalyses of the BIA

Stepped analysis

Scenario

Year 0 (current situation)

Year 1

Year 2

Year 3

Three-year total

Sponsor’s corrected base case

Reference

$2,617,101

$4,639,396

$5,144,813

$5,274,895

$15,059,105

New drug

$2,617,101

$4,921,707

$7,220,907

$8,878,228

$21,020,842

Budget impact

$0

$282,311

$2,076,094

$3,603,333

$5,961,737

CADTH base case

Reference

$3,047,404

$5,433,975

$6,044,313

$6,206,471

$17,684,758

New drug

$3,047,404

$5,739,188

$8,114,429

$9,241,878

$23,095,495

Budget impact

$0

$305,213

$2,070,116

$3,035,408

$5,410,737

CADTH scenario analysis (revised base case with lowered BV market share)

Reference

$2,697,682

$4,827,655

$5,376,156

$5,526,829

$15,730,640

New drug

$2,697,682

$5,346,450

$7,927,420

$9,113,897

$22,387,767

Budget impact

$0

$518,795

$2,551,263

$3,587,068

$6,657,127

BIA = budget impact analysis.

Note: This analysis is based on the publicly available prices of the comparator treatments.