Replication of Overall Survival, Progression-Free Survival, and Overall Response in Chemotherapy Arms of Non-Small Cell Lung Cancer Trials Using Real-World Data

Thanh G N Ton, Navdeep Pal, Huong Trinh, Sami Mahrus, Michael T Bretscher, Robson J M Machado, Natalia Sadetsky, Nayan Chaudhary, Michael W Lu, Gregory J Riely, Thanh G N Ton, Navdeep Pal, Huong Trinh, Sami Mahrus, Michael T Bretscher, Robson J M Machado, Natalia Sadetsky, Nayan Chaudhary, Michael W Lu, Gregory J Riely

Abstract

Purpose: The utility of real-world data (RWD) for use as external controls in drug development is informed by studies that replicate trial control arms for different endpoints. The purpose of this study was to replicate control arms from four non-small cell lung cancer (NSCLC) randomized controlled trials (RCT) to analyze overall survival (OS), progression-free survival (PFS), and overall response rate (ORR) using RWD.

Patients and methods: This study used RWD from a nationwide de-identified database and a clinico-genomic database to replicate OS, PFS, and ORR endpoints in the chemotherapy control arms of four first-line NSCLC RCTs evaluating atezolizumab [IMpower150-wild-type (WT), IMpower130-WT, IMpower131, and IMpower132]. Additional objectives were to develop a definition of real-world PFS (rwPFS) and to evaluate the real-world response rate (rwRR) endpoint.

Results: Baseline demographic and clinical characteristics were balanced after application of propensity score weighting methods. For rwPFS and OS, RWD external controls were generally similar to their RCT control counterparts. Across all four trials, the hazard ratio (HR) point estimates comparing trial controls with external controls were closer to 1.0 for the PFS endpoint than for the OS endpoint. An exploratory assessment of rwRR in RWD revealed a slight but nonsignificant overestimation of RCT ORR, which was unconfounded by baseline characteristics.

Conclusions: RWD can be used to reasonably replicate the OS and PFS of chemotherapy control arms of first-line NSCLC RCTs. Additional studies can provide greater insight into the utility of RWD in drug development.

Trial registration: ClinicalTrials.gov NCT02366143 NCT02367781 NCT02367794 NCT02657434.

©2022 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Adjustment of baseline characteristics between the RCT and RWD arms for OS and rwPFS assessments. Adjustments of characteristics for IMpower150-WT (A), IMpower130-WT (B), IMpower131 (C), and IMpower132 (D) where red indicates raw data, green indicates data with trimming, and blue indicates data after weighting and trimming. Standardized mortality ratio with no trimming was applied to patients in the RCT. For final analysis, inverse probability weighting with average treatment effect for the treated population estimates with no trimming were applied to the RCT cohort to achieve cohort balancing. Ideal balance occurs when the data have SMD < 0.1 (bold line) for all baseline characteristics. BEV, bevacizumab; C, carboplatin; EC, external control; nabP; nab-paclitaxel; P, paclitaxel; PEM, pemetrexed; Plat, carboplatin/cisplatin. *, Duration was defined as the time from initial diagnosis to index date.
Figure 2.
Figure 2.
Primary results to replicate PFS of RCT control arms using RWD. Kaplan–Meier curves comparing PFS for the RCT control arms versus the RWD EC arms in the IMpower150-WT (A), IMpower130-WT (B), IMpower131 (C), and IMpower132 RCTs (D). Standard mortality ratio weighting without trimming was performed. Sample sizes may differ from OS, as some patients lacked follow-up data extending beyond baseline. EC, external control. *, WT intent-to-treat population without epidermal growth factor/anaplastic lymphoma kinase alterations.
Figure 3.
Figure 3.
Primary results to replicate OS of RCT control arms using RWD. Kaplan–Meier curves comparing OS for the RCT control arms versus the RWD EC arms in the IMpower150-WT (A), IMpower130-WT (B), IMpower131 (C), and IMpower132 RCTs (D). Standard mortality ratio weighting without trimming was performed. Sample sizes may differ from PFS, as some patients lacked follow-up data extending beyond baseline. EC, external control. *, WT intent-to-treat population without epidermal growth factor/anaplastic lymphoma kinase alterations.
Figure 4.
Figure 4.
Replication of PFS and OS of control arms of IMpower RCTs using RWD. HRs of PFS (A) and OS (B) of RCT control versus RWD EC arms in the IMpower150-WT, IMpower130-WT, IMpower131, and IMpower132 RCTs. EC, external control. *, Reference group is the RCT control arm.
Figure 5.
Figure 5.
RCT ORR and unadjusted rwRR according to restrictive criteria group. rwRR of RCT control versus RWD EC arms in the IMpower150-WT (A), IMpower130-WT (B), IMpower131 (C), and IMpower132 RCTs (D). EC, external control; ITT, intent to treat.

References

    1. Khozin S, Blumenthal GM, Pazdur R. Real-world data for clinical evidence generation in oncology. J Natl Cancer Inst 2017;109.
    1. . H.R.34 - 21st Century Cures Act. Available from: .
    1. US Food and Drug Administration. Framework for FDA's real-world evidence program. 2018. Available from: .
    1. Arondekar B, Duh MS, Bhak RH, DerSarkissian M, Huynh L, Wang K, et al. . Real-world evidence in support of oncology product registration: a systematic review of new drug application and biologics license application approvals from 2015–2020. Clin Cancer Res 2022;28:27–35.
    1. Wedam S, Fashoyin-Aje L, Bloomquist E, Tang S, Sridhara R, Goldberg KB, et al. . FDA approval summary: palbociclib for male patients with metastatic breast cancer. Clin Cancer Res 2020;26:1208–12.
    1. Friends of Cancer Research. A blueprint for breakthrough: exploring utility of real-world evidence (RWE). Available from: .
    1. Carrigan G, Whipple S, Capra WB, Taylor MD, Brown JS, Lu M, et al. . Using electronic health records to derive control arms for early phase single-arm lung cancer trials: proof-of-concept in randomized controlled trials. Clin Pharmacol Ther 2020;107:369–77.
    1. Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, et al. . Atezolizumab for first-line treatment of metastatic non-squamous NSCLC. N Engl J Med 2018;378:2288–301.
    1. West H, McCleod M, Hussein M, Morabito A, Rittmeyer A, Conter HJ, et al. . Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non–small cell lung cancer (IMpower130): a multicenter, randomized, open-label, phase III trial. Lancet Oncol 2019;20:924–37.
    1. Jotte R, Cappuzzo F, Vynnychenko I, Stroyakovskiy D, Rodríguez-Abreu D, Hussein M, et al. . Atezolizumab in combination with carboplatin and nab-paclitaxel in advanced squamous NSCLC (IMpower131): results from a randomized phase III trial. J Thorac Oncol 2020;15:1351–60.
    1. Papadimitrakopoulou V, Cobo M, Bordoni R, Dubray-Longeras P, Szalai Z, Ursol G, et al. . OA05.07 IMpower132: PFS and safety results with 1L atezolizumab + carboplatin/cisplatin + pemetrexed in stage IV non-squamous NSCLC. J Thorac Oncol 2018;13:S332–S3.
    1. Ma X, Long L, Moon S, Adamson BJS, Baxi SS. Comparison of population characteristics in real-world clinical oncology databases in the US: Flatiron Health, SEER, and NPCR. medRxiv 2020.
    1. Birnbaum B, Nussbaum N, Seidle-Rathkopf K, Agrawal M, Estevez M, Estola E, et al. . Model-assisted cohort selection with bias analysis for generating large-scale cohorts from the EHR for oncology research. arXiv 2020.
    1. Singal G, Miller PG, Agarwala V, Li G, Kaushik G, Backenroth D, et al. . Association of patient characteristics and tumor genomics with clinical outcomes among patients with non–small cell lung cancer using a clinicogenomic database. JAMA 2019;321:1391–9.
    1. Curtis MD, Griffith SD, Tucker M, Taylor MD, Capra WB, Carrigan G, et al. . Development and validation of a high-quality composite real-world mortality endpoint. Health Serv Res 2018;53:4460–76.
    1. Zhang Q, Gossai A, Monroe S, Nussbaum NC, Parrinello CM. Validation analysis of a composite real-world mortality endpoint for patients with cancer in the United States. Health Serv Res 2021;56:1281–7.
    1. Ma X, Bellomo L, Magee K, Bennette CS, Tymejczyk O, Samant M, et al. . Characterization of a real-world response variable and comparison with RECIST-based response rates from clinical trials in advanced NSCLC. Adv Ther 2021;38:1843–59.
    1. Brookhart MA, Wyss R, Layton JB, Stürmer T. Propensity score methods for confounding control in nonexperimental research. Circ Cardiovasc Qual Outcomes 2013;6:604–11.
    1. Tan K, Bryan J, Segal B, Bellomo L, Nussbaum N, Tucker M, et al. . Emulating control arms for cancer clinical trials using external cohorts created from electronic health record-derived real-world data. Clin Pharmacol Ther 2022;111:168–78.
    1. Stewart M, Norden AD, Dreyer N, Henk HJ, Abernethy AP, Chrischilles E, et al. . An exploratory analysis of real-world end points for assessing outcomes among immunotherapy-treated patients with advanced non–small cell lung cancer. JCO Clin Cancer Inform 2019;3:1–15.
    1. Griffith SD, Tucker M, Bowser B, Calkins G, Chang CJ, Guardino E, et al. . Generating real-world tumor burden endpoints from electronic health record data: comparison of RECIST, radiology-anchored, and clinician-anchored approaches for abstracting real-world progression in non–small cell lung cancer. Adv Ther 2019;36:2122–36.
    1. Huang Bartlett C, Mardekian J, Cotter MJ, Huang X, Zhang Z, Parrinello CM, et al. . Concordance of real-world versus conventional progression-free survival from a phase III trial of endocrine therapy as first-line treatment for metastatic breast cancer. PLoS One 2020;15:e0227256.
    1. Feinberg BA, Zettler ME, Klink AJ, Lee CH, Gajra A, Kish JK. Comparison of solid tumor treatment response observed in clinical practice with response reported in clinical trials. JAMA Netw Open 2021;4:e2036741.
    1. Feinberg BA, Bharmal M, Klink AJ, Nabhan C, Phatak H. Using Response Evaluation Criteria in Solid Tumors in real-world evidence cancer research. Future Oncol 2018;14:2841–8.
    1. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, et al. . New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 2000;92:205–16.
    1. Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, et al. . NCCN guidelines insights: non–small cell lung cancer, version 2.2021. J Natl Compr Canc Netw 2021;19:254–66.
    1. Adamson BJS, Ma X, Griffith SD, Sweeney EM, Sarkar S, Bourla AB. Differential frequency in imaging-based outcome measurement: bias in real-world oncology comparative-effectiveness studies. Pharmacoepidemiol Drug Saf 2022;31:46–54.
    1. Betensky RA. Measures of follow-up in time-to-event studies: Why provide them and what should they be? Clin Trials 2015;12:403–8.
    1. Signorovitch JE, Sikirica V, Erder MH, Xie J, Lu M, Hodgkins PS, et al. . Matching-adjusted indirect comparisons: a new tool for timely comparative effectiveness research. Value Health 2012;15:940–7.

Source: PubMed

3
Subskrybuj