Background
Lung cancer is the most common cancer worldwide1 and the second most common in the United States, with an estimated 228,820 new cases predicted for 2020.2 Non–small cell lung cancer (NSCLC) is the most common subtype, accounting for 84% of all new US lung cancer diagnoses.3 At diagnosis, approximately 30% of patients with NSCLC have locally advanced stage III disease, many of whom have unresectable tumors.4
Historically, the prognosis for patients with locally advanced, unresectable stage III NSCLC has been poor. After platinum-based doublet concurrent chemoradiotherapy (cCRT; the previous standard of care5), median progression-free survival (PFS) was approximately 8 months, and 5-year overall survival (OS) rates were approximately 15% to 30%.5–8 However, the development of immune checkpoint inhibitors such as durvalumab that target either the PD-1 receptor or its ligand (PD-L1) has revolutionized the outlook for these patients.
Durvalumab is a selective, high-affinity, human immunoglobulin G1 monoclonal antibody that binds to PD-L1, blocking its interaction with PD-1 in the tumor microenvironment.9–11 This immune “unmasking” enables T cells to recognize tumor cells and eliminate them.9–11 In the phase III PACIFIC study (ClinicalTrials.gov identifier: NCT02125461) performed in patients with locally advanced, unresectable stage III NSCLC whose disease had not progressed after cCRT (defined as ≥2 overlapping cycles of platinum-based chemoradiotherapy), durvalumab significantly prolonged PFS and OS versus placebo.12,13 Median PFS was 16.8 months with durvalumab and 5.6 months with placebo (stratified hazard ratio [HR], 0.52; 95% CI, 0.42–0.65; P<.001).13 Furthermore, median OS was not reached with durvalumab versus 28.7 months with placebo (stratified HR, 0.68; 95% CI, 0.53–0.87; P=.0025).12
In February 2018, the FDA approved durvalumab for patients with unresectable stage III NSCLC that has not progressed after platinum-based cCRT,14 and durvalumab is now approved in >40 countries. Adjuvant therapy with durvalumab (also known as “consolidation immunotherapy”) after treatment with cCRT in patients with unresectable stage III NSCLC is also recommended in the 2018 (category 2A),15 2019 (category 1),16 and 2020 (category 1) NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for NSCLC.17 It is therefore important to determine whether durvalumab following cCRT is a cost-effective strategy.
We performed a cost-effectiveness analysis of durvalumab following cCRT versus cCRT alone in patients with locally advanced, unresectable stage III NSCLC, modeled from a US healthcare perspective, using data from the PACIFIC study.12 The primary objective was to determine the incremental cost-effectiveness ratio (ICER) of durvalumab, expressed as the cost per quality-adjusted life-year (QALY) gained for durvalumab following cCRT versus cCRT alone, using a $100,000 willingness-to-pay (WTP) threshold.
Methods
Patient Population
Modeling was based on the PACIFIC intention-to-treat population, comprising patients with locally advanced, unresectable stage III NSCLC that had not progressed after platinum-based cCRT. The intention-to-treat population was predominantly male (70% in both the durvalumab and placebo arms) and had a median age of 64 years in each arm and a mean weight of 71.1 kg overall. In the model, this information was used together with US general population mortality data18 to ensure that at no time point did modeled patients have greater expected survival than members of the general population.
Model Structure
A 3-state semi-Markov model was used to compare the 2 different treatment strategies from Medicare and commercial insurance perspectives. The 3 health states were progression-free, progressed disease, and death (Figure 1).

Three-state semi-Markov model structure.
Abbreviations: OS, overall survival; PFS, progression-free survival; PPS, postprogression survival; TTP, time to progression.
Citation: Journal of the National Comprehensive Cancer Network 19, 2; 10.6004/jnccn.2020.7621

Three-state semi-Markov model structure.
Abbreviations: OS, overall survival; PFS, progression-free survival; PPS, postprogression survival; TTP, time to progression.
Citation: Journal of the National Comprehensive Cancer Network 19, 2; 10.6004/jnccn.2020.7621
Three-state semi-Markov model structure.
Abbreviations: OS, overall survival; PFS, progression-free survival; PPS, postprogression survival; TTP, time to progression.
Citation: Journal of the National Comprehensive Cancer Network 19, 2; 10.6004/jnccn.2020.7621
PFS, time to progression (TTP), and post progression survival (PPS) data from PACIFIC (data cutoff [DCO], March 22, 2018) were used to generate probabilities that patients remained in the same health state or transitioned to a subsequent health state. These probabilities were sufficient to estimate OS at each cycle (Figure 1). Due to the relative immaturity of OS data from the March 22, 2018 DCO and the later January 31, 2019 DCO, too much uncertainty would have been introduced into the model if a partitioned survival approach (using only PFS and OS data) had been used; this approach was therefore used only for a scenario analysis.
Because PPS was similar between the durvalumab and placebo arms for the first 13 months of the study, with only a slight separation in favor of durvalumab thereafter, PPS data from both arms were pooled, and a conservative assumption was made of no difference in PPS between arms. To extrapolate beyond the trial follow-up period, parametric survival curves were fitted separately to the durvalumab and best supportive care arms using patient-level data from the PACIFIC trial.12,13
All fitted parametric curves were compared and assessed using the following goodness-of-fit criteria:
• Akaike information criterion (AIC) and Bayesian information criterion (BIC) with smaller AIC/BIC values indicating a better statistical fit. In general, models with a difference in AIC and BIC <5 were assumed to be of equal statistical fit.
• A visual inspection of the fitted curves: the fitted parametric survival model curves were overlaid on the Kaplan-Meier curves to assess how closely the modeled curves matched the observed nonparametric survival estimates.
• An external validation process through PACIFIC survival data and the wider clinical literature: the modeled median OS and PFS and the survival prob abilities at years 1, 2, 3, and 5 were compared with the reported median and survival estimates from the PACIFIC,12 START,19 and RTOG 016720 trial data.
A time horizon of 30 years was adopted, reflecting a lifetime approach capturing all costs and outcomes. Cycle length was 2 weeks for the first 12 months (to align with durvalumab administration in the PACIFIC trial, whereby patients received treatment until disease progression or for up to 12 months), and 4 weeks thereafter (to decrease computational time).
Efficacy
Effectiveness inputs were based on PACIFIC PFS data from the March 22, 2018 DCO. A sensitivity analysis was also performed using immature OS data from the January 31, 2019 DCO. PFS was defined as the time from randomization until objective disease progression or death from any cause in the absence of progression.12 TTP was defined as the time from randomization to progression, censoring death events. PPS was defined as the time from progression to death from any cause. Because PPS was similar between the durvalumab and placebo arms for the first 13 months of the PACIFIC study, with only a slight separation in favor of durvalumab thereafter (supplemental eFigure 1, available with this article at JNCCN.org), PPS data from both treatment arms were pooled.
For each treatment arm, standard parametric distributions (exponential, Weibull, log-normal, log-logistic, Gompertz, and generalized gamma) were fitted to the Kaplan-Meier curves for PFS (assessed by blinded independent central review [BICR]), PPS (BICR), and TTP. The best-fitting curves were selected according to the lowest Akaike/Bayesian information criteria and best visual fit. The generalized gamma curve was deemed the best fitting for PFS and TTP, and the log-logistic curve was considered the best fitting for PPS. Because the duration of preprogression treatment effect for durvalumab is unknown, a conservative assumption of no further benefit beyond 60 months was made, after which treatment effect was assumed to be the same as for cCRT alone; this duration was varied in the scenario analyses. PPS was pooled across both arms, and an assumption of no difference was made. Efficacy outputs (ie, QALYs and LYs gained) were assumed to be the same for both Medicare and commercial insurance.
Utility
In the PACIFIC trial, health-related quality of life data were collected using the EuroQoL 5-dimension 5-level (EQ-5D-5L) questionnaire.21 In the model, EQ-5D-5L data were converted to US population–based utility values using published algorithms.22,23
Adverse effect (AE)–related disutilities were not considered in the base-case analysis, because the impact of AEs on health-related quality of life was assumed to be accounted for already in patients’ health state utilities. There were also only minor differences in AE rates between arms in PACIFIC; thus, their impact on model outcomes was considered only minimal.
Costs
A full breakdown of modeled costs is provided in supplemental eTables 1–5. Unit drug costs were sourced from the Centers for Medicare & Medicaid Services Average Sales Price Drug Pricing Files (October 201824; “Medicare”) and wholesale acquisition costs from the IBM Micromedex RED BOOK25 (“commercial insurance”). All costs were standardized to 2018 US dollars and, when required, were inflated using the US Bureau of Labor Statistics Consumer Price Index health inflation indices.26 Drug costs were calculated assuming no vial sharing for durvalumab, consolidation with chemotherapy, or subsequent therapy. Time to treatment discontinuation was used to inform total drug acquisition and treatment administration costs. In PACIFIC, all patients were discontinued from treatment at or before 12 months per protocol; mean times to treatment discontinuation were 7.7 and 6.9 months for durvalumab and placebo, respectively.
Consolidation with chemotherapy was applied as a one-off cost and comprised paclitaxel + carboplatin (4 cycles) or pemetrexed + cisplatin (2 cycles) (supplemental eTable 1). To reflect real-world treatment of unresectable stage III NSCLC in the United States, 36.2% of patients in the cCRT-alone arm were assumed to receive subsequent consolidation with chemotherapy (paclitaxel + carboplatin, 33.9%; pemetrexed + cisplatin, 2.2%).27 However, studies have shown that consolidation with chemotherapy does not improve survival28; thus, it was assumed that these patients incurred no survival advantage.
Costs associated with healthcare resource use (HRU) were estimated retrospectively from the IBM MarketScan Commercial Claims and Encounters Database.29 Patients with unresectable stage III NSCLC who underwent cCRT were identified from the database and observed until 3 months post progression (proxied by initiation of subsequent therapy line). Monthly reimbursed costs for their HRU, including outpatient oncologist visits and radiology scans in the pre and post progression periods, were calculated (supplemental eTable 2).
One line of subsequent (post progression) treatment was accounted for in the model, and included immunotherapy (nivolumab or pembrolizumab, per proportions of patients receiving these as subsequent therapies in the PACIFIC study12; supplemental eTable 6), cytotoxic therapy, and radiotherapy (supplemental eTables 3 and 4). For clarity, we note that the NCCN Guidelines for NSCLC do not recommend subsequent immunotherapy after progression on durvalumab or PD-1/PD-L1 inhibitors.17 Inclusion of further lines was expected to have only a minor impact on model outputs.
The model also included costs associated with grade 3 or 4 AEs occurring in ≥2% of patients in either arm of the PACIFIC study (supplemental eTable 5). End-of-life care costs were applied as a one-off cost and assumed to be the same in both arms.
Discounting
Costs and QALYs were discounted at a rate of 3% per annum30; rates of 0% and 5% per annum were explored as scenario analyses.
Sensitivity Analyses
One-way sensitivity analyses investigated the effects of individual model parameters on ICER. Costs and treatment durations were varied using a gamma distribution whereby utilities and the proportions of patients receiving each treatment were varied using a beta distribution, assuming a standard error (SE) of 10% of the base-case value. When data relating to uncertainty were not available, the lower and upper bounds were varied using the 2.5 and 97.5 percentiles, assuming a 10% SE.
A probabilistic sensitivity analysis (PSA) was performed to explore the joint uncertainty of all model parameters and their associated impact on cost-effectiveness, and to quantify the level of confidence in the model outputs. When data were available, variation was based on actual data; when unavailable, a 10% SE was assumed. The PSA was run 1,000 times. The proportion of iterations considered cost-effective was determined and represented via a cost-effectiveness acceptability curve. Details of scenario analyses and model validation are shown in supplemental eAppendix 1.
Results
Base-Case Analysis
Discounted results for durvalumab following cCRT versus cCRT alone are presented in Table 1. Durvalumab following cCRT was associated with an incremental increase of 1.94 LYs and 1.65 QALYs versus cCRT alone. Incremental total costs were $91,423 for Medicare and $101,058 for commercial insurance, yielding ICERs of $55,285 and $61,111 per QALY gained, respectively, which were both below the $100,000 WTP threshold.
Discounted Incremental Cost-Effectiveness of Durvalumab (Base-Case Analysis)


Durvalumab following cCRT was associated with a greater increase in LYs and QALYs gained in the progression-free state (+2.37 and +2.01, respectively) but fewer LYs and QALYs gained in the progressed-disease state (−0.43 and −0.36, respectively) versus cCRT alone, because fewer patients in the durvalumab arm entered the post progression state (when durvalumab/placebo therapy was no longer being administered) during the modeled time horizon.
The relatively high drug and administration costs associated with durvalumab were partly offset by lower discounted end-of-life and subsequent therapy costs. Durvalumab was also associated with higher HRU costs because patients spent more time in the progression-free health state. A full breakdown of discounted and undiscounted costs is detailed in supplemental eTables 7 and 8.
One-Way Sensitivity Analyses
From the Medicare perspective, no variable increased the ICER above the $100,000 WTP threshold, and all variables had only a minor impact (Figure 2A). Variables with the biggest impact on the ICER were the proportion of patients receiving nivolumab subsequent therapy (cCRT-alone arm) and the duration of nivolumab treatment (cCRT-alone arm). Similar results were observed for commercial insurance (Figure 2B).

One-way sensitivity analysis examining the impact of individual model parameters on the ICER of durvalumab following cCRT versus cCRT alone, from (A) Medicare and (B) commercial insurance perspectives. Note: nivolumab use as subsequent treatment was based on post progression therapy from the PACIFIC trial.12
Abbreviations: BSC, best supportive care; cCRT, concurrent chemoradiotherapy; freq, frequency; ICER, incremental cost-effectiveness ratio; mono, monotherapy; PFS, progression-free survival; QALY, quality-adjusted life-year.
Citation: Journal of the National Comprehensive Cancer Network 19, 2; 10.6004/jnccn.2020.7621

One-way sensitivity analysis examining the impact of individual model parameters on the ICER of durvalumab following cCRT versus cCRT alone, from (A) Medicare and (B) commercial insurance perspectives. Note: nivolumab use as subsequent treatment was based on post progression therapy from the PACIFIC trial.12
Abbreviations: BSC, best supportive care; cCRT, concurrent chemoradiotherapy; freq, frequency; ICER, incremental cost-effectiveness ratio; mono, monotherapy; PFS, progression-free survival; QALY, quality-adjusted life-year.
Citation: Journal of the National Comprehensive Cancer Network 19, 2; 10.6004/jnccn.2020.7621
One-way sensitivity analysis examining the impact of individual model parameters on the ICER of durvalumab following cCRT versus cCRT alone, from (A) Medicare and (B) commercial insurance perspectives. Note: nivolumab use as subsequent treatment was based on post progression therapy from the PACIFIC trial.12
Abbreviations: BSC, best supportive care; cCRT, concurrent chemoradiotherapy; freq, frequency; ICER, incremental cost-effectiveness ratio; mono, monotherapy; PFS, progression-free survival; QALY, quality-adjusted life-year.
Citation: Journal of the National Comprehensive Cancer Network 19, 2; 10.6004/jnccn.2020.7621
PSA
Results of the PSA agreed with those of the base-case analysis from both payer perspectives (Table 2). The probability that durvalumab following cCRT was cost-effective versus cCRT alone, with an ICER <$100,000, was approximately 97.4% for Medicare and 97.3% for commercial insurance (Figure 3).
Costs, QALYs, and ICERs Generated From Probabilistic Sensitivity and Base-Case Analyses



Cost-effectiveness acceptability curves for the durvalumab following cCRT and cCRT-alone arms generated from the probabilistic sensitivity analysis (1,000 iterations) from (A) Medicare and (B) commercial insurance perspectives.
Abbreviations: cCRT, concurrent chemoradiotherapy; WTP, willingness to pay.
Citation: Journal of the National Comprehensive Cancer Network 19, 2; 10.6004/jnccn.2020.7621

Cost-effectiveness acceptability curves for the durvalumab following cCRT and cCRT-alone arms generated from the probabilistic sensitivity analysis (1,000 iterations) from (A) Medicare and (B) commercial insurance perspectives.
Abbreviations: cCRT, concurrent chemoradiotherapy; WTP, willingness to pay.
Citation: Journal of the National Comprehensive Cancer Network 19, 2; 10.6004/jnccn.2020.7621
Cost-effectiveness acceptability curves for the durvalumab following cCRT and cCRT-alone arms generated from the probabilistic sensitivity analysis (1,000 iterations) from (A) Medicare and (B) commercial insurance perspectives.
Abbreviations: cCRT, concurrent chemoradiotherapy; WTP, willingness to pay.
Citation: Journal of the National Comprehensive Cancer Network 19, 2; 10.6004/jnccn.2020.7621
Scenario Analyses
Alternative assumptions that contributed to model uncertainty included time horizon ($59,616–$83,106 for Medicare and $65,206–$87,634 for commercial insurance), duration of treatment effect ($44,529–$66,114 and $50,664–$71,628, respectively), choice of fitted PFS curve ($42,570–$60,889 and $48,768–$66,562, respectively), duration of subsequent immunotherapy ($54,810–$61,606 and $60,630–$67,518, respectively), and use of a partitioned survival model ($58,145 and $64,016, respectively). However, no scenario increased the ICER above the $100,000 WTP threshold. Full details of all scenarios are presented in Table 3.
Scenario Analyses From Medicare and Commercial Insurance Perspectives


Model Validation
Validation results for modeled PFS and OS data are presented in supplemental eTable 9 and eFigure 2.
Discussion
Durvalumab following cCRT was cost-effective compared with cCRT alone at a WTP threshold of $100,000 from Medicare (ICER, $55,285 per QALY gained) and commercial insurance ($61,111 per QALY gained) perspectives. The $100,000 WTP threshold was selected because it was at the lower end of the range recommended by Neumann et al.31 However, a threshold of $150,000 may also be considered acceptable for cancer drugs,32 including NSCLC,33 meaning the ICER of durvalumab also fell well below the upper theoretical WTP threshold for new oncologic interventions.
Sensitivity analyses showed the robustness of the model results and revealed that the cost-effectiveness of durvalumab was particularly sensitive to use and duration of subsequent (ie, post progression) immunotherapy. In all PSAs, durvalumab following cCRT was cost-effective compared with cCRT alone, and scenario analyses showed that the model was most sensitive to assumptions regarding time horizon, duration of durvalumab treatment effect, choice of fitted PFS curve, use of a partitioned survival model structure, and duration of subsequent immunotherapy. However, the ICER of durvalumab remained below $100,000 in all 3 scenarios (3, 6, and 12 months of subsequent immunotherapy). Although we acknowledge that the NCCN Guidelines for NSCLC do not recommend subsequent immunotherapy after progression on durvalumab or PD-1/PD-L1 inhibitors,17 this sensitivity analysis directly addresses potential bias toward durvalumab and instead endeavors to better reflect real-world clinical care.
Despite durvalumab being associated with lower incremental costs, shorter time horizon and duration of treatment effect increased the ICER because fewer incremental LYs/QALYs were gained in these scenarios. Likewise, the ICER changed when using the alternative partitioned-survival model, whereby the ICER increased to $58,145 to $64,106. This was because of the reduction in incremental LYs/QALYs accrued, which may be explained by the fact that, as of the March 22, 2018 DCO, the PFS curve had plateaued, suggesting long-term benefits from cCRT and/or durvalumab. However, there were too few deaths to produce a plateau in the OS curve. Consequently, the extrapolated PFS and OS curves crossed each other, which is theoretically implausible. Because the partitioned-survival model relies on direct extrapolation of these curves, their crossing will have biased results.
Results from our base-case analysis agree with those published by Criss et al,34 who used a decision analytic microsimulation model to determine the cost-effectiveness and potential budgetary impacts of durvalumab following cCRT in patients with NSCLC. Among 2 million simulated patients, the ICER of durvalumab following cCRT versus cCRT alone was $67,421 per QALY, showing cost-effectiveness at the $100,000 WTP threshold.34 The investigators concluded that giving patients durvalumab earlier in the treatment course, when the goal of treatment is to prolong survival, potentially with curative intent, and before patients develop metastatic disease, would provide a cost-effective means of prolonging survival.34 However, a number of key differences between our analysis and that of Criss et al34 should be noted. First, durvalumab data (PFS) in the Criss model were based on less-mature PFS data from the first data readout, and OS data were unavailable,34 whereas the data used in our analysis were from the more recent data readout of the PACIFIC trial with approximately 2 years’ longer follow-up. Second, we were able to use the actual average sales pricing for durvalumab because the treatment is now in use, whereas Criss et al34 used an estimate for this. Third, we were able to include second-line immunotherapy in our model, although we acknowledge that despite subsequent immunotherapy not being included in the original model by Criss et al,34,35 when added in a later analysis, durvalumab remained cost-effective, with an ICER of $79,609 per QALY. Therefore, our analysis builds on the work of Criss et al35 to provide a more robust estimate of the costs associated with durvalumab consolidation therapy, given current clinical practice patterns.
Economic evaluations are central to the process by which payers in countries such as Australia, Canada, and members of the European Union assess the value of new interventions.36 In the United States, moves toward adopting value-based frameworks have begun to emerge, primarily due to escalating healthcare costs, particularly in cancer care.37–39 As the level of importance placed on drug value has increased in the United States, institutions and medical professional societies have responded, developing new frameworks to assess the value of novel interventions. In oncology, these include the ASCO Value Framework Net Health Benefit score,40 NCCN Evidence Blocks,41 NCCN Framework for Resource Stratification of NCCN Guidelines (NCCN Framework),42 ESMO Magnitude of Clinical Benefit Scale,43 Institute for Clinical and Economic Review Value Assessment Framework,44 and Memorial Sloan Kettering Cancer Center Drug Abacus.45 In addition to evaluating the efficacy and safety of an intervention, these frameworks account for other factors underlying value, such as evidence quality, impact on quality of life, and cost-effectiveness.38,39 Consistent with the goal of these frameworks to evaluate the value of oncologic treatments, our study found durvalumab following cCRT to be cost-effective compared with cCRT alone in patients with unresectable stage III NSCLC, showing the long-term value of durvalumab in this setting.
The main limitation of the state-transition model used in our analysis was that PFS, TTP, and PPS data were immature; therefore, these curves had to be extrapolated, resulting in model uncertainty (although extrapolated data were validated clinically). However, the state-transition model offers several advantages over models used more frequently in oncology cost-effectiveness analyses, such as partitioned-survival models, in which survival curves for PFS and OS are fitted completely independently from each other. As described earlier, if PFS and OS data are of different maturity, the more mature endpoint may exhibit trends different from those of the less-mature endpoint, potentially producing contradictory results when the survival curves are combined. This effect is accentuated when a long survival tail is expected, such as in stage III NSCLC, in which 9.4% to 11.6% of patients remain alive and progression-free at 5 years after CRT.7
Conclusions
Our findings show that in a US healthcare setting, durvalumab following cCRT is cost-effective compared with cCRT alone, supporting the adoption of this regimen as the new standard of care in unresectable stage III NSCLC.
Acknowledgments
The authors thank Peter Elroy (BresMed Health Solutions, Sheffield, UK) for the global model development, and Hannah Kilvert and Tim Grant (BresMed Health Solutions, Sheffield, UK) for the utility analyses. Medical writing support during the preparation of the manuscript, which was in accordance with Good Publication Practice (GPP3) guidelines, was provided by Catherine Crookes of Cirrus Communications (Macclesfield, UK), an Ashfield company.
References
- 1.↑
WHO. Cancer. September 12, 2018. Accessed February 27, 2019. Available at: https://www.who.int/news-room/fact-sheets/detail/cancer
- 2.↑
American Cancer Society. Key statistics for lung cancer. Accessed October 12, 2020. Available at: https://www.cancer.org/cancer/non-small-cell-lung-cancer/about/key-statistics.html
- 3.↑
Cancer.Net. Lung cancer – non-small cell: statistics. Accessed October 12, 2020. Available at: https://www.cancer.net/cancer-types/lung-cancer-non-small-cell/statistics
- 4.↑
Chen VW, Ruiz BA, Hsieh MC, et al.. Analysis of stage and clinical/prognostic factors for lung cancer from SEER registries: AJCC staging and collaborative stage data collection system. Cancer 2014;120(Suppl 23):3781–3792.
- 5.↑
Yoon SM, Shaikh T, Hallman M. Therapeutic management options for stage III non-small cell lung cancer. World J Clin Oncol 2017;8:1–20.
- 6.↑
Ahn JS, Ahn YC, Kim JH, et al.. Multinational randomized phase III trial with or without consolidation chemotherapy using docetaxel and cisplatin after concurrent chemoradiation in inoperable stage III non-small-cell lung cancer: KCSG-LU05-04. J Clin Oncol 2015;33:2660–2666.
- 7.↑
Aupérin A, Le Péchoux C, Rolland E, et al.. Meta-analysis of concomitant versus sequential radiochemotherapy in locally advanced non-small-cell lung cancer. J Clin Oncol 2010;28:2181–2190.
- 8.↑
Bradley JD, Hu C, Komaki R, et al.. Long-term results of RTOG 0617: a randomized phase 3 comparison of standard dose versus high dose conformal chemoradiation therapy +/− cetuximab for stage III NSCLC [abstract]. Int J Radiat Oncol Biol Phys 2017;99(Suppl 2):S105.
- 9.↑
Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252–264.
- 10.↑
Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol 2015;33:1974–1982.
- 11.↑
Stewart R, Morrow M, Hammond SA, et al.. Identification and characterization of MEDI4736, an antagonistic anti-PD-L1 monoclonal antibody. Cancer Immunol Res 2015;3:1052–1062.
- 12.↑
Antonia SJ, Villegas A, Daniel D, et al.. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med 2018;379:2342–2350.
- 13.↑
Antonia SJ, Villegas A, Daniel D, et al.. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med 2017;377:1919–1929.
- 14.↑
Durvalumab. Prescribing information. AstraZeneca; 2018. Accessed February 27, 2019. Available at: https://www.azpicentral.com/imfinzi/imfinzi.pdf#page=1
- 15.↑
Bironzo P, Di Maio M. A review of guidelines for lung cancer. J Thorac Dis 2018;10(Suppl 13):S1556–1563.
- 16.↑
Besette Z. Guideline updates: NCCN extensively updates guideline for NSCLC. Journal of Clinical Pathways, November 6, 2018. Accessed May 13, 2019. Available at: https://www.journalofclinicalpathways.com/news/nccn-extensively-updates-guideline-nsclc
- 18.↑
Social Security Administration. Actuarial life table: 2015. Accessed February 27, 2019. Available at: https://www.ssa.gov/oact/STATS/table4c6.html
- 19.↑
Butts C, Socinski MA, Mitchell PL, et al.. Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START): a randomised, double-blind, phase 3 trial. Lancet Oncol 2014;15:59–68.
- 20.↑
Bradley JD, Paulus R, Komaki R, et al.. Standard-dose versus high-dose conformal radiotherapy with concurrent and consolidation carboplatin plus paclitaxel with or without cetuximab for patients with stage IIIA or IIIB non-small-cell lung cancer (RTOG 0617): a randomised, two-by-two factorial phase 3 study. Lancet Oncol 2015;16:187–199.
- 21.↑
Herdman M, Gudex C, Lloyd A, et al.. Development and preliminary testing of the new five-level version of EQ-5D (EQ-5D-5L). Qual Life Res 2011;20:1727–1736.
- 22.↑
Shaw JW, Johnson JA, Coons SJ. US valuation of the EQ-5D health states: development and testing of the D1 valuation model. Med Care 2005;43:203–220.
- 23.↑
van Hout B, Janssen MF, Feng YS, et al.. Interim scoring for the EQ-5D-5L: mapping the EQ-5D-5L to EQ-5D-3L value sets. Value Health 2012;15:708–715.
- 24.↑
Centers for Medicare & Medicaid Services. 2018 ASP drug pricing files. Accessed February 27, 2019. Available at: https://www.cms.gov/Medicare/Medicare-Fee-for-Service-Part-B-Drugs/McrPartBDrugAvgSalesPrice/2018ASPFiles.html
- 25.↑
IBM Watson Health. IBM Micromedex RED BOOK. Accessed February 27, 2019. Available at: https://www.ibm.com/us-en/marketplace/micromedex-red-book
- 26.↑
U.S. Bureau of Labor Statistics. Consumer Price Index. Accessed February 27, 2019. Available at: https://www.bls.gov/cpi
- 27.↑
Witlox WJA, van Asselt ADI, Wolff R, et al.. Durvalumab for the treatment of locally advanced, unresectable, stage III non-small cell lung cancer: an evidence review group perspective of a NICE single technology appraisal. Pharmacoeconomics 2020;38:317–324.
- 28.↑
Tsujino K, Kurata T, Yamamoto S, et al.. Is consolidation chemotherapy after concurrent chemo-radiotherapy beneficial for patients with locally advanced non-small-cell lung cancer? A pooled analysis of the literature. J Thorac Oncol 2013;8:1181–1189.
- 29.↑
Adamson DM, Chang S, Hansen LG. White paper: health research data for the real world: the MarketScan databases. Accessed June 4, 2019. Available at: https://www.ibm.com/downloads/cas/6KNYVVQ2
- 30.↑
Sanders GD, Neumann PJ, Basu A, et al.. Recommendations for conduct, methodological practices, and reporting of cost-effectiveness analyses: second panel on cost-effectiveness in health and medicine. JAMA 2016;316:1093–1103.
- 31.↑
Neumann PJ, Cohen JT, Weinstein MC. Updating cost-effectiveness—the curious resilience of the $50,000-per-QALY threshold. N Engl J Med 2014;371:796–797.
- 32.↑
Bae YH, Mullins CD. Do value thresholds for oncology drugs differ from nononcology drugs? J Manag Care Spec Pharm 2014;20:1086–1092.
- 33.↑
Institute for Clinical and Economic Review. Treatment options for advanced non-small cell lung cancer: effectiveness, value and value-based price benchmarks. Accessed June 24, 2019. Available at: https://icer.org/wp-content/uploads/2020/10/MWCEPAC_NSCLC_Final_Evidence_Report_Meeting_Summary_110116.pdf
- 34.↑
Criss SD, Mooradian MJ, Sheehan DF, et al.. Cost-effectiveness and budgetary consequence analysis of durvalumab consolidation therapy vs no consolidation therapy after chemoradiotherapy in stage III non-small cell lung cancer in the context of the US health care system. JAMA Oncol 2019;5:358–365.
- 35.↑
Criss SD, Mooradian MJ, Kong CY. Changes to model assumptions of the cost-effectiveness of durvalumab therapy for non-small cell lung cancer—in reply. JAMA Oncol 2019;5:1066–1067.
- 36.↑
Jørgensen J, Servos S, Kefalas P. The potential price and access implications of the cost-utility and budget impact methodologies applied by NICE in England and ICER in the US for a novel gene therapy in Parkinson’s disease. J Mark Access Health Policy 2018;6:1500419.
- 37.↑
Mariotto AB, Yabroff KR, Shao Y, et al.. Projections of the cost of cancer care in the United States: 2010-2020. J Natl Cancer Inst 2011;103:117–128.
- 38.↑
Neumann PJ, Cohen JT. Measuring the value of prescription drugs. N Engl J Med 2015;373:2595–2597.
- 39.↑
Slomiany M, Madhavan P, Kuehn M, et al.. Value frameworks in oncology: comparative analysis and implications to the pharmaceutical industry. Am Health Drug Benefits 2017;10:253–260.
- 40.↑
Schnipper LE, Davidson NE, Wollins DS, et al.. Updating the American Society of Clinical Oncology value framework: revisions and reflections in response to comments received. J Clin Oncol 2016;34:2925–2934.
- 41.↑
National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) with NCCN Evidence Blocks. Accessed April 21, 2020. Available at: https://www.nccn.org/evidenceblocks/default.aspx
- 42.↑
National Comprehensive Cancer Network. NCCN Framework for Resource Stratification of NCCN Guidelines (NCCN Framework). Accessed April 21, 2020. Available at: https://www.nccn.org/framework/
- 43.↑
Cherny NI, Sullivan R, Dafni U, et al.. A standardised, generic, validated approach to stratify the magnitude of clinical benefit that can be anticipated from anti-cancer therapies: the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS). Ann Oncol 2015;26:1547–1573.
- 44.↑
Institute for Clinical and Economic Review. Overview of the ICER value assessment framework and update for 2017–2019. Accessed June 24, 2019. Available at: https://icer.org/wp-content/uploads/2020/10/ICER-value-assessment-framework-Updated-050818.pdf
- 45.↑
Memorial Sloan Kettering Cancer Center, Drug Pricing Laboratory. Drug Abacus. Accessed May 13, 2019. Available at: http://abacus.realendpoints.com/abacus-mskcc?ab-eff=1000&ab-tox=0.1&ab-nov=1&ab-rare=1&ab-pop=1&ab-dev=1&ab-prog=1.0&ab-need=1&ab-time=1607377695