Cost-effectiveness analysis of pembrolizumab versus chemotherapy as the first-line treatment in patients with mismatch-repair-deficient (dMMR) or microsatellite- instability-high (MSI-H) advanced or metastatic colorectal cancer from a perspective of health care system in China

DOI: https://doi.org/10.21203/rs.3.rs-2079558/v1

Abstract

Background

Pembrolizumab is superior to chemotherapy as a first-line treatment for patients with mismatch-repair-deficient (dMMR) or microsatellite-instability-high (MSI-H) advanced or metastatic colorectal cancer (CRC), with a significantly long-term survival benefit trend by KYENOTE-177. The current study aimed to determine whether pembrolizumab is a cost-effective treatment for patients with dMMR/MSI-H advanced or metastatic CRC in China.

Methods

A partitioned survival model (PSM) was developed to simulate patients with dMMR/MSI-H advanced or metastatic CRC based on health states of progression-free survival (PFS), progressive disease (PD) and death. The model was designed using a lifetime horizon, a 6-week cycle, and a 5% discount rate. The characteristics of patients in the model were similar to patients in a phase 3, open-label randomized clinical trial (KEYNOTE-177) who had metastatic MSI-H–dMMR CRC and had not previously received treatment. The health outcomes and utilities were from the KETNOTE-177 trial and published data, respectively. Costs were calculated based on local charges (2021) and published literature. It is deemed cost-effective in China if the incremental cost-effectiveness ratio (ICER) value is less than $33,600 per quality adjusted life-year (QALY). The robustness of the results was discussed in one-way deterministic and probabilistic sensitivity analyses.

Results

Baseline analysis disclosed that pembrolizumab provided an additional 2.58 QALYs (3.00 life-years) at an incremental cost of $78,286.04, resulting in an ICER of $30,330.15 per QALY, which was below the willingness-to-pay threshold of $33,600 per QALY. When patient assistance program (PAP) was considered, the ICER became $1,730.67 per QALY, manifesting absolute cost-effectiveness. The results of sensitivity analyses demonstrated that in most cases pembrolizumab was cost-effective.

Conclusions

Pembrolizumab is a cost-effective first-line treatment for dMMR/MSI-H advanced or metastatic CRC patients in China, especially considering PAP.

1. Introduction

Colorectal cancer (CRC) is one of the most common malignant tumors in China, ranking second in the overall cancer incidence and first in the digestive tract cancer[1]. In 2020, about 550,000 people were diagnosed with CRC and about 280,000 died of CRC, which is second only to lung cancer in China[2]. Besides, the economic burden of CRC continued to grow. The average annual growth rate of the medical expenditure per CRC patient in China ranged from 6.9–9.2%, and the 1-year out-of-pocket expenditure of a newly diagnosed patient accounted for about 60% of their previous-year household income.[3–8]

Meanwhile, more than half of CRC patients were diagnosed as advanced stage (III - IV)[9, 10] with poor prognosis and distant metastasis, leaving 5-year overall survival (OS) rate < 15%[11]. For these patients, standard chemotherapy are 5-fluorouracil (5-FU) based regimens, such as FOLFOX (5-FU, oxaliplatin, and leucovorin) or FOLFIRI (5-FU, irinotecan, and leucovorin) alone or in combination with therapies that block epidermal growth factor receptor (EGFR) or vascular endothelial growth factor (VEGF) signaling.[12] However, the long-term efficacy of standard chemotherapy is generally poor because that among the patients with advanced CRC, DNA mismatch repair defect (dMMR) /microsatellite instability high (MSI-H) account for 5%-10%, which is associated with resistance to standard chemotherapy and a poor prognosis.[13, 14] Fortunately, with the rapid development of immune checkpoint inhibitors (ICIs) over the past decades, several elegant clinical trials (KEYNOTE-016[15], KEYNOTE-164[16], KEYNOTE-177[17], CheckMate-142[18]) have shown that programmed death 1 (PD-1) blockade have achieved durable responses in patients with dMMR/MSI-H metastatic CRC (mCRC) that are refractory to standard chemotherapy combinations.[16, 18–21]

Pembrolizumab was the first PD-1 inhibitor approved by the National Medical Products Administration (NMPA) in China to be adopted as the standard of care first-line treatment in patients with dMMR/MSI-H mCRC based on the data analysis results of a key global phase III clinical trial - KEYNOTE-177[17]. Recently, 2021 American Society of Clinical Oncology (ASCO) shown an exciting result that at a median follow-up of more than 44 months (44.5 [36.0-60.3] months in the pembrolizumab group and 44.4 [36.2–58.6] months in the control group), the median OS were not yet achieved (pembrolizumab group) and 36.7 months (control group) respectively, and the risk of death was reduced by 26% (HR, 0.74, 95% CI [0.53–1.03]; P = 0.0359). Although 60% of the patients in the control group (i.e., doublet chemotherapy ± targeted therapy: either FOLFOX or FOLFIRI, with or without either cetuximab or bevacizumab) were interfered by PD-1/programmed death-ligands1(PD-L1) ICIs after disease progression, pembrolizumab group still showed a long-term OS benefit trend. At 36 months, 61% of the patients in the pembrolizumab group were still alive, which was 11% higher than that in the control group. In the second interim analysis of KEYNOTE-177[17], pembrolizumab was found to significantly improve the progression-free survival (PFS), compared to the control group, as a first-line treatment for dMMR/MSI-H mCRC (median PFS, 16.5 vs. 8.2 months; HR, 0.60; P = 0.0002). Superior PFS in the pembrolizumab group, remained consistent in subgroup analyses of age, sex, race, region, stage, and BRAF status.

Despite the exciting incremental benefit, relative higher cost without national medical insurance of pembrolizumab makes the value of this therapy relative to its benefit remains indeterminate. To settle this issue, we built a partitioned-survival model (PSM) to evaluate the cost-effectiveness of pembrolizumab as first-line regime in patients with dMMR/MSI-H mCRC from a perspectives of health care system in China with a lifetime horizon.

2. Materials And Methods

2.1 Model structure

To compare the cost-effectiveness of pembrolizumab versus chemotherapy in patients with MSI-H–dMMR stage IV CRC, a PSM was conducted to simulate the process of dMMR/MSI-H mCRC depending on the clinical data from KEYNOTE 177. Three distinct health states: PFS, progressive disease (PD) and death (Fig. 1) were included in the model. The initial state is assumed to be PFS, and death is the absorbing state. The population was a cohort with the same characteristics and treatments as those in the KEYNOTE 177 trial. Unlike Markov model, the PSM use of the trials' Kaplan-Meier (K-M) curves to directly divide patients into different health states without putting forward assumptions for the transition of patients between different health states. Therefore, the estimation of patients’ proportion in each health state was acquired straightly from the cumulative survival probabilities in OS and PFS curves by parametric functions fitting and extrapolation. The cycle length was 6-week. The analysis was conducted from the Chinese health care system perspective with a life-time horizon so as to ensure there were less than 1% survivors. Costs, life years (LYs), quality-adjusted life years (QALYs), and incremental cost-effectiveness ratios (ICERs) were calculated in each treatment group. If the ICER is below $33,600 threshold (three times GDP per capita of China in 2020, ¥210,000.00), the treatment is generally considered to be cost-effective. In line with Chinese pharmacoeconomic guidelines[22], both costs and benefits are discounted at 5% (range: 0%-8%) per year.

2.2 Efficacy Estimates

We used the Engauge Digitizer (version 12.1, http://digitizer.sourceforge.net) to collect the data points from the K-M curves (PFS and OS curves) of the two arms and followed the method of Guyot et al.[23] to reconstruct estimates of underlying individual patient data (IPD) over the clinical trial time. In terms of the IPD out of the clinical trial time, standard parametric models fitting and extrapolation were used to estimate the long-term survival probabilities by using R software (version 4.1.0, https://www.r-project.org). Specifically, six parametric functions were considered, including exponential, Weibull, Gompertz, log-logistic, log-normal, and generalized gamma distributions. And then, in order to evaluate the goodness-of-fit of each parametric survival model, multiple methods were applied such as visual inspection, Akaike information criterion (AIC) test and Bayesian information criteria (BIC) test proposed by NICE DSU technical support document 14 (TSD14)[24]. Lower AIC and BIC values indicate better fit of the selected parametric model. (eTable 1–4 are given in Online Resource) Meanwhile, superposed graphs of the K-M curves from the trial and the estimated curves based on the relative better fitting parametric survival models were presented in Fig. 2 so as to intuitively inspect the survival prediction.

The generalized gamma model was chosen as the best fit model for the OS curve of both arms and PFS of pembrolizumab arm and log-normal model for the PFS curve of chemotherapy arm. Considerations were as follows: 1) the lowest or relative lower AIC and BIC values among all survival models; 2) the best fit with the observed curves based on visual inspection.

2.3 Utility Estimates

The health utility score reflects the level of physical, mental, and social functioning associated with a disease correlative health state that varies from 0 to 1, with 0 representing the worst health state/death and 1 representing the best. Average health utility of the PFS state was 0.7825, which was based on quality-of-life data collected in the KEYNOTE 177 trial.[25] In the trial, EQ-5D-3L index[26] mean utility scores were 0.77 in the pembrolizumab group and 0.75 in the chemotherapy group at baseline. At the end of week 18, mean scores were 0.84 and 0.77 respectively. So, in the simulation, we assigned a utility of 0.7825 for PFS, which is the mean of above values, with 0.75 and 0.84 as the boundaries of the range used in sensitivity analyses. Average health utility of the PD state was 0.64 (95% CI [0.576–0.704]) derived from the previously published literature[27]. The disutility values of grade 3–4 adverse events (AEs) were considered in our analysis,[28, 29] but only one-time assessment was carried out during the first cycle for simplification given the trivial influence of AE disutilities. QALYs loss caused by AEs were assessed by the product of the incidence of AEs and the corresponding disutility value. And ± 20% were the boundaries of the range in sensitivity analyses.

2.4 Cost Inputs

Only direct medical costs were considered, including cost of the drug utilization, cost of drug administration, cost of follow up, cost of main AEs management and cost of treatments for progression (including active treatments and supportive care), and stated in 2021 United States dollars (USD) by exchange rate: 1 CYN = 0.16 USD. Drug prices were estimated from the local bid-winning price.[30]

Drug administration costs were calculated as a function of administration cost per attendance and administration frequency (number of attendance per cycle). Unit administration costs were derived from local charge. The costs of follow up, including carcinoembryonic antigen level test, ultrasound of abdomen, outpatient specialist clinic, were from a published study[31], and were converted to 2020 USD using Medical Care component of Chinese Consumer Price Index (CPI). The frequency of follow up were based on the guideline of Chinese society of clinical oncology (CSCO) for colorectal cancer[12]. The costs of main AEs management were derived from the previously published literatures[32, 33] (adjusted by CPI) and were calculated only once in the first cycle. In terms of the treatments for progression, active therapeutic schedules included CAPIRI, CAPOX, FOLFOX (± bevacizumab or cetuximab), FOLFIRI (± bevacizumab or cetuximab), nivolumab, pembrolizumab.[34, 35] The prices of the drugs in subsequent treatments were also from local bid-winning price[30], and the fee of supportive care was assumed to be 0. Additionally, costs were discounted at an annual rate of 5%[22].

2.5 Clinical Inputs

According to the global phase III clinical trial - KEYNOTE-177[17], the patients in the pembrolizumab group (P group, N = 153) received pembrolizumab at a dose of 200 mg every 3 weeks intravenously (IV) for up to 35 treatments (approximately 2 years) and the patients in the chemotherapy group (C group, N = 154) received FOLFOX (oxaliplatin 85 mg/m2 IV on Day 1, leucovorin 400 mg/m2 IV on Day 1, 5-FU 400 mg/m2 IV bolus on Day 1 and then 1200 mg/m2/day IV over 2 days for total dose of 2400 mg/m2 in each 2-week cycle) or FOLFIRI (irinotecan 180 mg/m2 IV on Day 1, leucovorin 400 mg/m2 IV on Day 1, 5-FU 400 mg/m2 IV bolus on Day 1 and then 1200 mg/m2/day IV over 2 days for total dose of 2400 mg/m2 in each 2-week cycle), with or without either cetuximab (cetuximab 400 mg/m2 IV over 2 hours then 250 mg/m2 over 1 hour weekly in each 2-week cycle) or bevacizumab (5 mg/kg IV on Day 1 of each 2-week cycle). Treatment was continued until disease progression, development of unacceptable toxic effects, illness, or a decision by the physician or patient to withdraw from the trial. In C group, the percentage of patients in mFOLFOX, mFOLFOX + bevacizumab, mFOLFOX + cetuximab, FOLFIRI, FOLFIRI + bevacizumab and FOLFIRI + cetuximab were 7.69%, 44.76%, 3.50%, 11.19%, 25.17% and 7.69%, respectively. After disease progression, patients randomly assigned to the C group could cross over to pembrolizumab (to receive a maximum of 35 treatments) and patients in the P group can continue pembrolizumab (to receive a maximum of 17 treatments). The subsequent treatments were mainly composed of pembrolizumab (P group 10%, C group 42.75%), other PD-1/PD-L1 ICIs (P group 7.5%, C group 28.24%), chemotherapy (P group 43.75%, C group 15.27%), VEGF inhibitor (P group 27.5%, C group 9.92%), and EGFR (P group 11.25%, C group 3.82%) inhibitor. Referring to the other studies and guideline[34, 35], we assumed that CAPIRI, CAPOX, FOLFOX and FOLFIRI were used as the standard second-line chemotherapy, nivolumab (up to 2 years) and pembrolizumab as the ICIs, cetuximab as the EGFR inhibitor and bevacizumab as the VEGF inhibitor, which are commonly used in China. We included grade 3 to 4 AEs in the model that had obvious clinical impact and significantly different rates between the arms of the KEYNOTE 177 trial, which were diarrhea, anemia, hypokalemia, and neutropenia. The incidence of neutrophil count decrease multiplied by 0.5 is included in neutropenia. For dosage calculation, assuming that values of body surface area (BSA) and weight were 1.80 m2 and 65kg, respectively.[36]

2.6 Sensitivity Analysis

A series of sensitivity analyses were conducted to test the robustness of the model and address uncertainty in the estimation of model parameters by using Microsoft Excel (version 16.51). One-way deterministic sensitivity analyses (DSA) were used to evaluate the impact of uncertainty of a single input variable on the ICER. The range of drug prices depends on the local charge or ± 20% of the baseline values. Other parameters were adjusted within the reported 95% confidence intervals (CI) or assuming reasonable ranges of the base case values (± 20%) if 95% CIs were unavailable, in accordance with established approach[37]. In addition, a separate scenario considering patient assistance program (PAP) was evaluated in the DSA.

In the probabilistic sensitivity analysis (PSA), a Monte Carlo simulation of 500 iterations was generated by randomly sampling the key model parameters from the pre-specified distributions simultaneously. We used gamma distribution for the cost parameters and beta distribution for utility and probability parameters. Based on the data from 500 iterations, a cost-effectiveness acceptability curve (CEAC) was created to represent the likelihood that pembrolizumab would be considered cost-effective compared with chemotherapy on the basis of a willingness to pay (WTP) threshold of $33,600 per QALY in China.

Baseline values, ranges, and distributions for sensitivity analysis were summarized in Table 1

Table 1

Baseline values, ranges, and distributions for sensitivity analysis

Variable

Baseline value

Range

α

β

mean

SE

Reference for baseline value

Distribution (parameters)

Minimum

Maximum

Cycle

6-week

-

-

-

-

-

-

-

-

Horizon

Life-time

-

-

-

-

-

-

-

-

WTP, $/QALY

33600

-

-

-

-

-

-

[21]

-

Discount rate

0.05

0

0.08

-

-

-

-

[21]

-

Mean body surface area, m2

1.80

-

-

-

-

-

-

-

-

Patients’ weight, kg

65

-

-

-

-

-

-

-

-

Pembrolizumab group _ AEs incidence

Diarrhea

0.060

0.048

0.072

90.218

1413.409

0.060

0.006

[16]

Beta

Anemia

0.050

0.040

0.060

91.188

1732.572

0.050

0.005

Beta

Hypokalemia

0.010

0.008

0.012

95.070

9411.890

0.010

0.001

Beta

Neutropenia

0.000

0.000

0.000

-

-

0.000

0.000

Beta

Chemotherapy group _ AEs incidence

Diarrhea

0.110

0.088

0.132

85.366

690.685

0.110

0.011

[16]

Assumed the incidence of neutrophil count decrease multiplied by 0.5 is included in neutropenia

Beta

Anemia

0.100

0.080

0.120

86.336

777.024

0.100

0.010

Beta

Hypokalemia

0.060

0.048

0.072

90.218

1413.409

0.060

0.006

Beta

Neutropenia

0.235

0.188

0.282

73.236

238.405

0.235

0.024

Beta

Pembrolizumab group second-line therapy proportion

Pembrolizumab

0.100

0.080

0.120

86.336

777.024

0.100

0.010

KEYNOTE 177, assumed only the second-line treatments of a proportion of more than 2% were considered in our analysis.

Beta

other PD-1/PD-L1 Checkpoint Inhibitor

0.075

0.060

0.090

88.762

1094.731

0.075

0.008

Beta

Chemotherapy

0.438

0.350

0.525

53.585

68.895

0.438

0.045

Beta

VEGF inhibitor

0.275

0.220

0.330

69.354

182.842

0.275

0.028

Beta

EGFR Inhibitor

0.113

0.090

0.135

85.123

671.526

0.113

0.011

Beta

Chemotherapy group second-line therapy proportion

Pembrolizumab

0.427

0.342

0.513

54.557

73.068

0.427

0.044

KEYNOTE 177, assumed only the second-line treatments of a proportion of more than 2% were considered in our analysis.

Beta

other PD-1/PD-L1 Checkpoint Inhibitor

0.282

0.226

0.339

68.632

174.362

0.282

0.029

Beta

Chemotherapy

0.153

0.122

0.183

81.225

450.797

0.153

0.016

Beta

VEGF inhibitor

0.099

0.079

0.119

86.410

784.338

0.099

0.010

Beta

EGFR Inhibitor

0.038

0.031

0.046

92.336

2326.872

0.038

0.004

Beta

Health preferences

Utility of PFS

0.7825

0.75

0.84

245.001

63.176

0.795

0.023

[24]

Beta

Utility of PD

0.64

0.576

0.704

137.658

77.432

0.640

0.033

[26]

Beta

Disutility due to AEs (grade ≥ 3)

Diarrhea

-0.090

-0.072

-0.108

14.470

146.308

-0.090

0.023

[27][28]

Beta

Anemia

-0.085

-0.068

-0.102

14.555

156.680

-0.085

0.021

Beta

Hypokalemia

-0.080

-0.064

-0.096

14.640

168.360

-0.080

0.020

Beta

Neutropenia

-0.0607

-0.049

-0.073

14.968

231.623

-0.061

0.015

Beta

Drug cost, $/mg

Pembrolizumab

179.18

-

-

-

-

-

-

2021 Local charge

https://www.yaozh.com

Fix

Oxaliplatin

3.4

2.72

4.08

16.000

0.213

3.400

0.850

Gamma

Leucovorin

0.25

0.2

0.3

16.000

0.016

0.250

0.063

Gamma

5-FU

0.29

0.232

0.348

16.000

0.018

0.290

0.073

Gamma

Bevacizumab

15

11.88

15

16.000

0.840

13.440

3.360

Gamma

Cetuximab

12.04

-

-

-

-

-

-

Fix

Irinotecan

17.73

14.05

24.9

16.000

1.217

19.475

4.869

Gamma

Nivolumab

96.2

-

-

-

-

-

-

Fix

Capecitabine

0.04

0.01

0.04

16.000

0.002

0.025

0.006

Gamma

AEs cost, $

Diarrhea

392.44

313.95

470.93

16.000

24.527

392.439

98.110

[32]

Gamma

Anemia

724.64

579.71

869.57

16.000

45.290

724.638

181.160

[31]

Gamma

Hypokalemia

157.28

125.82

188.73

16.000

9.830

157.275

39.319

[31], assumed same as fatigue

Gamma

Neutropenia

628.96

503.17

754.76

16.000

39.310

628.965

157.241

[31]

Gamma

Administration, $/attendance

310.16

248.13

372.19

16.000

19.385

310.160

77.540

Local charge

Gamma

Cost of BSC, $/cycle

0.00

0.00

0.00

-

-

-

-

Assumed to be 0

-

Follow up, $/cycle

31.33

25.06

37.60

16.000

1.958

31.330

7.833

[11][30]

Gamma

PD-1, programmed death 1; PD-L1, programmed death ligand 1; EGFR, epidermal growth factor receptor; VEGF, vascular endothelial growth factor; PD, progressive disease; PFS, progression-free survival; AE, adverse event; BSC, best supportive care.

3. Results

3.1 Base Case Results

The base case model results are listed in Table 2. Over a lifetime horizon, the use of pembrolizumab compared with chemotherapy produced a gain of extra 3.00 LYs. When adjusted for quality of life, the total QALYs for pembrolizumab and chemotherapy were estimated to be 6.71 and 4.13, respectively. According to the dosage scheme used, the PSM assessed the total costs to be $229,698.49 and $151,412.44, respectively, during that period, resulting in an ICER of $30,330.15 per QALY.

Table 2

Base-case results

Strategy

Chemotherapy

Pembrolizumab

Cost ($)

151,412.44

229,698.49

LYs

6.22

9.22

QALYs

4.13

6.71

Incremental cost a

NA

78,286.04

Incremental LYs a

NA

3.00

Incremental QALYs a

NA

2.58

Incremental Cost per LY Gained a

NA

$8,284.72

Incremental Cost per QALY Gained a

NA

$30,330.15

LYs, life years; QALYs, quality-adjusted life years.
a Compared with chemotherapy

3.2 Sensitivity Analysis

3.2.1 Deterministic sensitivity analysis results

The results of DSA are presented in the tornado diagram (Fig. 3). The parameters with the greatest influence on the ICER were consideration of PAP, discount rate for cost and effectiveness, proportion of subsequent treatments, and baseline utility value. Across arbitrary change in the ranges for each parameter, the ICER remained ༜$33,600.00 per QALY, except that the discount rate became 0.08 and the proportion of VEGF inhibitor used as 2nd line regime in pembrolizumab group increased by 20%. The incidence, cost, and disutility for AEs had a tiny impact on the ICER. Besides, when PAP was under consideration, the ICER became $1,730.67 per QALY, manifesting absolute cost-effectiveness.

3.2.2 Probabilistic sensitivity analysis results

The results of the PSA are shown in the CEAC in Fig. 4, which show the probability that pembrolizumab is cost-effective along with increase of WTP values. These outcomes indicated nearly 0% probability that pembrolizumab was cost-effective at WTP values $20,000 per QALY. There were a 50% and 100% opportunity that pembrolizumab was cost-effective at WTP values of approximately $30,000 and $40,000 per QALY, respectively. The scatter plot shown in Fig. 5 depict the results of the 500 simulations of the PSA, in which, most results were under the WTP threshold ($33,600 per QALY), indicating that probably pembrolizumab generated more QALYs with acceptable incremental costs.

4. Discussion

4.1 Summary and Interpretation of Results

A 5-FU in combination with either oxaliplatin or irinotecan (± VEGF or EGFR inhibitor) are established treatments for mCRC, with good clinical efficacy at an acceptable cost.[15, 38–46] With the dramatic development of ICIs over the past decades, some clinical trials[15–18] have indicated that PD-1 blockade used alone, such as pembrolizumab and nivolumab approved by NMPA or FDA, have achieved much more effectiveness in patients with dMMR/MSI-H metastatic or unresectable CRC that are resistant to the standard chemotherapy combinations on a longer course of treatment.[16, 18–21] A study[47] suggested that increased tumor grading (captured by Ki-67) is associated with impairment of anti‐tumor immunity through HLA-I down modulation, reduced CD8 infiltration and enhanced PD-L1/PD-1 expression on tumor cells. Therefore, it may be possible to predict the efficacy of ICIs by detecting the expression of Ki-67. However, these PD-1 blockade are with significant cost, and their value remains under judgment. Cost-effectiveness analyses provide a feasible methodology for evaluating the cost-effectiveness of a regimen, under the circumstances of the survival benefit, quality of life, costs of administration and drugs, costs of follow up and costs of AEs. But at present, there exist no cost-effectiveness analysis of pembrolizumab or nivolumab used alone in the patients with dMMR/MSI-H mCRC.

Pembrolizumab was the first and only one PD-1 inhibitor approved by the NMPA in China as the first-line therapy for patients with dMMR/MSI-H mCRC based on the gratifying results of KEYNOTE 177 trial. The trial demonstrated that by the treatment of pembrolizumab, survival could be prolonged significantly compared with standard chemotherapy. In the Chinese context, given the incremental benefit and cost related to this treatment, we conducted the first study to our knowledge detecting the cost-effectiveness of pembrolizumab, as the first-line regimen, in patients with dMMR/MSI-H advanced or metastatic CRC. On the basis of our model, pembrolizumab was projected to extend patient life expectancy to a point not previously seen in this patient population. For instance, in the pembrolizumab arm, life expectancy was projected to reach to nearly 9 years, approximately 1.5 times that of the chemotherapy group (increase patients’ survival by 3 years with discounting), despite the high proportion of crossover in the control group after disease progression. The associated incremental cost per QALY gained was $30,330.15. While there is no single established ICER threshold for cost-effectiveness in China, the World Health Organization has referenced a threshold of 3-times estimated per capita GDP with respect to disability-adjusted life years, which for China for 2020 would correspond to $33,600/QALY.[48] The ICER in the basic analysis is below this threshold, suggesting pembrolizumab monotherapy could be selected as a cost-effective first-line treatment for patients with dMMR/MSI-H advanced or metastatic CRC in clinical practice.

In univariable sensitivity analyses, the most influential driver of the ICER was the consideration of PAP. The PAP called “Key to life” was approved in China on July 8, 2021[49], officially providing support that alleviates patients’ out-of-pocket expenses of pembrolizumab (as the first-line monotherapy) for dMMR/MSI-H metastatic or unresectable CRC patients with wild-type KRAS, NRAS and BRAF genes. Based on this assistance program, it only takes $11,200 per year (up to 2 years, $22,400) with a decrease of about 88.8%. Considering that, we conducted a scenario analysis and found that the basic ICER became $1,730.67 per QALY, which indicated that the patients could receive more benefits with a minimal incremental costs. Under such circumstance, we fully recommend the mCRC patients, who are meet the above-mentioned conditions, to use pembrolizumab monotherapy as the first-line regime. The impact of discount rate on the basic results ranked second. In addition, due to the relatively high price of biological agents[30], the proportion of VEGF/EGFR inhibitor used as 2nd line regimes also had a great effect on the outcomes. For this, some scholars[50, 51] have found that treatment with bevacizumab in Chinese patients with mCRC is unlikely to use financial resources efficiently. In terms of EGFR inhibitor (cetuximab), Wu, B. et al.[52] and Wang, H. et al.[53] have conducted cost-effective analyses based on the CRYSTAL trial[54] and the TAILOR trial[55] respectively, and found that cetuximab combined with FOLFOX or FOLFIRI was not a cost-effective treatment for the patients with RAS wild-type mCRC in China, unless PAP was available. The PSA, varying all model parameters in 500 Monte Carlo simulations, displayed that it was more than 80% probability that pembrolizumab would be considered cost-effective at a usually accepted value ($33,600 per QALY).

4.2 Strengths and Limitations

A major strength of the basic analysis was its dependence on a direct comparison of pembrolizumab and standard chemotherapy, utilizing data and information from a randomized controlled trial. Besides, the PSM structure is unnecessary to build assumptions for the transition probabilities of patients but has the advantage of being able to partition patients to different health states directly based on the trial's K-M curves. Moreover, time dependence of risk can be handled automatically, instead of applying constant transition risks within a traditional Markov model.

It is essential to address that our study had several limitations. Firstly, the development of the cost-effective analysis was relied on the results of the KEYNOTE 177 trial, in which the participants were mainly from the Western Europe or North America (༞70%). To some extent, it may impair the application of our research results. Secondly, the value of utilities of PFS and PD were derived from the study on the health-related quality of life in the trial population, and the disutilities of AEs were derived from previously published studies, which might not reflect the health state of patients in China. This defect may also affect the robustness of our results. An updated health utility data for patients with dMMR/MSI-H metastatic or unresectable CRC in Chinese populations might enhance the accuracy and robustness of the analyses .Thirdly, the efficacy data of PFS and the incidence of AEs were from the interim results of the trial, because the final results have not been officially published. Thus, it is possible that projections of cost-effectiveness could change with additional follow-up. Fourthly, without considering patient compliance, the cost of follow-up may be overestimated. Because of this, cost of follow-up as a variable was included in the DSA and we found that the impact of the follow-up cost on the results was small. In addition, for the longer-term extrapolation of PFS and OS, there existed an inherent uncertainty, and the perspective of Chinese health care system may limit the scope of application of the results.

5. Conclusion

From a perspective of health care system in China, the current model predicted that pembrolizumab monotherapy is more likely to be a cost-effective first-line strategy for patients with dMMR/MSI-H advanced or metastatic CRC. When PAP is available, pembrolizumab monotherapy will be dominant. The reported conclusions may be helpful to physicians, mCRC patients and health management agency in their decision-making processes.

List Of Abbreviations

colorectal cancer (CRC)

overall survival (OS)

5-fluorouracil (5-FU)

FOLFOX (5-FU, oxaliplatin, and leucovorin)

FOLFIRI (5-FU, irinotecan, and leucovorin)

epidermal growth factor receptor (EGFR)

vascular endothelial growth factor (VEGF)

mismatch repair defect (dMMR) /microsatellite instability high (MSI-H)

immune checkpoint inhibitors (ICIs)

programmed death 1 (PD-1)

programmed death-ligands1(PD-L1)

metastatic CRC (mCRC)

National Medical Products Administration (NMPA)

American Society of Clinical Oncology (ASCO)

progression-free survival (PFS)

partitioned-survival model (PSM)

progressive disease (PD)

Kaplan-Meier (K-M)

life years (LYs)

quality-adjusted life years (QALYs)

incremental cost-effectiveness ratios (ICERs)

individual patient data (IPD)

Akaike information criterion (AIC)

Bayesian information criteria (BIC)

adverse events (AEs)

United States dollars (USD)

Consumer Price Index (CPI)

Chinese society of clinical oncology (CSCO)

intravenously (IV)

body surface area (BSA)

deterministic sensitivity analyses (DSA)

confidence intervals (CI)

patient assistance program (PAP)

probabilistic sensitivity analysis (PSA)

cost-effectiveness acceptability curve (CEAC)

willingness to pay (WTP)

Declarations

Acknowledgments

No one need to be acknowledged.

Author Contributions: Drs Wu and Han had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. All authors read and approved the final manuscript.

Concept and design: Wu, Han.

Acquisition, analysis, or interpretation of data: All authors.

Drafting of the manuscript: Zhu, Han.

Critical revision of the manuscript for important intellectual content: All authors.

Statistical analysis: Zhu, Wu.

Administrative, technical, or material support: Wu, Han.

Supervision: Wu.

Notes

Ethics approval and consent to participate

This economic analysis was based on a literature review and modeling techniques; this study did not require approval from an Institutional Research Ethics Board.

Consent for publication

Not applicable.

Competing interests

None of the authors have anything to declare.

Funding

This work was sponsored by specific grant No. 2022ZYJ07 for pharmacoeconomics and health technology assessment from Zhejiang Pharmaceutical Association.

Data availability statement 

Most data generated or analysed during this study are included in this published article [and its supplementary information files]. Complete datasets are available from the corresponding author on reasonable request.

References

  1. Diseases GBD, Injuries C. Global burden of 369 diseases and injuries in 204 countries and territories, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019[J]. Lancet 2020, 396(10258):1204-1222.
  2. https://gco.iarc.fr/.
  3. Chang S BY, Pu HQ, et al. Disease burden of colorectal cancer in Jinchang cohor[J]. Chin J Epidemiol 2016, 37(3):325-328.
  4. Huang H, Shi, JF., Guo, LW. et al. Expenditure and financial burden for the diagnosis and treatment of colorectal cancer in China:a hospital-based, multicenter, cross-sectional survey[J]. Chin J Cancer 2017, 36(1):41.
  5. Liu CC HH, Shi JF, et al. Economic burden of colorectal cancer in China from 1996 to 2015:a systematic review[J]. China Cancer 2017, 26(11):859-867.
  6. Wang H, Cao MD, Liu CC, et al. Disease burden of colorectal cancer in China: any changes in recent years?[J]. Chinese Journal of Epidemiology 2020, 41(10):1633-1642.
  7. Y S. Study on the economic burden of malignant tumor in a Certain area of Southwest China. Tangshan: North China University of Science and Technology, 2019.
  8. Cai Y XM, Chen WQ, et al. Expenditure of hospital care on cancer in China, from 2011 to 2015[J]. Chin J Cancer Res 2017, 29(3):253-262.
  9. Lee YH, Kung PT, Wang YH, et al. Effect of length of time from diagnosis to treatment on colorectal cancer survival: A population-based study[J]. PLoS One 2019, 14(1):e0210465.
  10. Li X, Zhou Y, Luo Z, et al. The impact of screening on the survival of colorectal cancer in Shanghai, China: a population based study[J]. BMC Public Health 2019, 19(1):1016.
  11. Howlader N, Noone AM,Krapcho M, et al. SEER Cancer Statistics Revirw, 1975-2016 [DB/OL]. Bethesda (MD): National Cancer Institute, 2019 [2021-08-26]. https://seer.cancer.gov/csr/1975_2016.
  12. (CSCO) GOCSOCO. COLORECTAL CANCER 2018 V1. Accessed 26 Aug 2021.
  13. National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology. Colon Cancer. Version 1. 2020 [EB / OL]. Fort Washington (PA): National Comprehensive Cancer Network. 2019 [2021-08-26]. https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf.
  14. Lochhead P, Kuchiba A, Imamura Y, et al. Microsatellite instability and BRAF mutation testing in colorectal cancer prognostication[J]. J Natl Cancer Inst 2013, 105(15):1151-1156.
  15. Le DT, Uram JN, Wang H, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency[J]. N Engl J Med 2015, 372(26):2509-2520.
  16. Le DT, Kim TW, Van Cutsem E, et al. Phase II Open-Label Study of Pembrolizumab in Treatment-Refractory, Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic Colorectal Cancer: KEYNOTE-164[J]. J Clin Oncol 2020, 38(1):11-19.
  17. Andre T, Shiu KK, Kim TW, et al. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer[J]. N Engl J Med 2020, 383(23):2207-2218.
  18. Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study[J]. Lancet Oncol 2017, 18(9):1182-1191.
  19. Overman MJ, Lonardi S, Wong KYM, et al. Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer[J]. J Clin Oncol 2018, 36(8):773-779.
  20. Ganesh K, Stadler ZK, Cercek A, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential[J]. Nat Rev Gastroenterol Hepatol 2019, 16(6):361-375.
  21. Wu T, Wu X, Wang HY, et al. Immune contexture defined by single cell technology for prognosis prediction and immunotherapy guidance in cancer[J]. Cancer Commun (Lond) 2019, 39(1):21.
  22. Liu G, Hu S, Wu J, et al. China guidelines for pharmacoeconomic evaluations, 2020.
  23. Guyot P, Ades AE, Ouwens MJ, et al. Enhanced secondary analysis of survival data: reconstructing the data from published Kaplan-Meier survival curves[J]. BMC Med Res Methodol 2012, 129.
  24. National Institute for Health and Care Excellence (NICE). Technical support document 14. Survival analysis for economic evaluations alongside clinical trials - extrapolation with patient-level data. 2013; http://nicedsu.org.uk/wp-content/uploads/2016/03/NICE-DSU-TSD-Survival-analysis.updated-March-2013.v2.pdf. Accessed 27 Aug 2021.
  25. Andre T, Amonkar M, Norquist JM, et al. Health-related quality of life in patients with microsatellite instability-high or mismatch repair deficient metastatic colorectal cancer treated with first-line pembrolizumab versus chemotherapy (KEYNOTE-177): an open-label, randomised, phase 3 trial[J]. Lancet Oncol 2021, 22(5):665-677.
  26. EuroQol Research Foundation. EQ-5D-3L user guide, version 6.0. December, 2018. https://euroqol.org/publications/user-guides/. Accessed 27 Aug, 2021.
  27. Tikhonova IA, Huxley N, Snowsill T, et al. Economic Analysis of First-Line Treatment with Cetuximab or Panitumumab for RAS Wild-Type Metastatic Colorectal Cancer in England[J]. Pharmacoeconomics 2018, 36(7):837-851.
  28. Freeman K, Connock M, Cummins E, et al. Fluorouracil plasma monitoring: systematic review and economic evaluation of the My5-FU assay for guiding dose adjustment in patients receiving fluorouracil chemotherapy by continuous infusion[J]. Health Technol Assess 2015, 19(91):1-321, v-vi.
  29. Harrow BS, Eaton CB, Roberts MB, et al. Health utilities associated with hemoglobin levels and blood loss in postmenopausal women: the Women's Health Initiative[J]. Value Health 2011, 14(4):555-563.
  30. Drugdataexpy: Marketing imformation, Local Bid-wining Price. https ://datayaozhcom.
  31. Wong CK, Lam CL, Poon JT, et al. Direct medical costs of care for Chinese patients with colorectal neoplasia: a health care service provider perspective[J]. J Eval Clin Pract 2012, 18(6):1203-1210.
  32. Wu B, Dong B, Xu Y, et al. Economic evaluation of first-line treatments for metastatic renal cell carcinoma: a cost-effectiveness analysis in a health resource-limited setting[J]. PLoS One 2012, 7(3):e32530.
  33. Wu B, Ye M, Chen H, et al. Costs of trastuzumab in combination with chemotherapy for HER2-positive advanced gastric or gastroesophageal junction cancer: an economic evaluation in the Chinese context[J]. Clin Ther 2012, 34(2):468-479.
  34. Shenwei Bian YL, Gordon G.Liu. Cost-Effectivness Analysis of Second-Line Chemotherapy Therapies For Metastatic Colorectal Cancer[J]. China Journal of Pharmaceutical Economics 2016, 11(2):7-10.
  35. Hospital Authority of National Health Commission of the People's Republic of China CSoO, Chinese Medical Association. Chinese protocol of diagnosis and treatment of colorectal cancer(2020 edition)[J]. Chinese Journal of Practical Surgery 2020, 40(6):601-625.
  36. Wu B, Chen H, Shen J, et al. Cost-effectiveness of adding rh-endostatin to first-line chemotherapy in patients with advanced non-small-cell lung cancer in China[J]. Clin Ther 2011, 33(10):1446-1455.
  37. Zhang Y, Baik SH, Fendrick AM, et al. Comparing local and regional variation in health care spending[J]. N Engl J Med 2012, 367(18):1724-1731.
  38. Giantonio BJ, Catalano PJ, Meropol NJ, et al. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200[J]. J Clin Oncol 2007, 25(12):1539-1544.
  39. Hochster HS, Hart LL, Ramanathan RK, et al. Safety and efficacy of oxaliplatin and fluoropyrimidine regimens with or without bevacizumab as first-line treatment of metastatic colorectal cancer: results of the TREE Study[J]. J Clin Oncol 2008, 26(21):3523-3529.
  40. Saltz LB, Clarke S, Diaz-Rubio E, et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study[J]. J Clin Oncol 2008, 26(12):2013-2019.
  41. Van Cutsem E, Kohne CH, Hitre E, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer[J]. N Engl J Med 2009, 360(14):1408-1417.
  42. Bokemeyer C, Bondarenko I, Hartmann JT, et al. Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: the OPUS study[J]. Ann Oncol 2011, 22(7):1535-1546.
  43. Van Cutsem E, Kohne CH, Lang I, et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status[J]. J Clin Oncol 2011, 29(15):2011-2019.
  44. Bennouna J, Sastre J, Arnold D, et al. Continuation of bevacizumab after first progression in metastatic colorectal cancer (ML18147): a randomised phase 3 trial[J]. Lancet Oncol 2013, 14(1):29-37.
  45. Heinemann V, von Weikersthal LF, Decker T, et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer (FIRE-3): a randomised, open-label, phase 3 trial[J]. Lancet Oncol 2014, 15(10):1065-1075.
  46. Goldstein DA, Chen Q, Ayer T, et al. First- and second-line bevacizumab in addition to chemotherapy for metastatic colorectal cancer: a United States-based cost-effectiveness analysis[J]. J Clin Oncol 2015, 33(10):1112-1118.
  47. Milione M, Miceli R, Barretta F, et al. Microenvironment and tumor inflammatory features improve prognostic prediction in gastro-entero-pancreatic neuroendocrine neoplasms[J]. J Pathol Clin Res 2019, 5(4):217-226.
  48. National Bureau Statistics. 2020 per capitagross domestic product (GDP. https://data.stats.gov.cn/search.htm?s=%E4%BA%BA%E5%9D%87GDP.
  49. CPHCF. Primary Health Care Foundation of China “Key to life” patient assistance program (PAP). http://smzy.ilvzhou.com, 2021.
  50. Han J, Xiao D, Tan C, et al. Cost-Effectiveness Analysis of First-Line FOLFIRI Combined With Cetuximab or Bevacizumab in Patients With RAS Wild-Type Left-Sided Metastatic Colorectal Cancer[J]. Cancer Control 2020, 27(1):1073274820902271.
  51. Zhang PF, Wen F, Zhou J, et al. Cost-effectiveness analysis of capecitabine plus bevacizumab versus capecitabine alone in elderly patients with previously untreated metastatic colorectal cancer from Chinese societal perspective[J]. Clin Transl Oncol 2020, 22(1):103-110.
  52. Wu B, Yao Y, Zhang K, et al. RAS testing and cetuximab treatment for metastatic colorectal cancer: a cost-effectiveness analysis in a setting with limited health resources[J]. Oncotarget 2017, 8(41):71164-71172.
  53. Wang H, Huang L, Gao P, et al. Cost-effectiveness analysis of cetuximab combined with chemotherapy as a first-line treatment for patients with RAS wild-type metastatic colorectal cancer based on the TAILOR trial[J]. BMJ Open 2020, 10(2):e030738.
  54. Van Cutsem E, Köhne CH, Hitre E, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer[J]. N Engl J Med 2009, 360(14):1408-1417.
  55. Qin S, Li J, Wang L, et al. Efficacy and Tolerability of First-Line Cetuximab Plus Leucovorin, Fluorouracil, and Oxaliplatin (FOLFOX-4) Versus FOLFOX-4 in Patients With RAS Wild-Type Metastatic Colorectal Cancer: The Open-Label, Randomized, Phase III TAILOR Trial[J]. J Clin Oncol 2018, 36(30):3031-3039.