Abstract
PURPOSE
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are standard first-line therapy for EGFR-mutant, metastatic non–small cell lung cancer (NSCLC); however, most patients experience disease progression. We report results from the randomized, double-blind, phase III KEYNOTE-789 study of pemetrexed and platinum–based chemotherapy with or without pembrolizumab for TKI-resistant, EGFR-mutant, metastatic nonsquamous NSCLC (ClinicalTrials.gov identifier: NCT03515837).
METHODS
Adults with pathologically confirmed stage IV nonsquamous NSCLC, documented DEL19 or L858R EGFR mutation, and progression after EGFR-TKI treatment were randomly assigned 1:1 to 35 cycles of pembrolizumab 200 mg or placebo once every 3 weeks plus four cycles of pemetrexed and carboplatin or cisplatin once every 3 weeks and then maintenance pemetrexed. Dual primary end points were progression-free survival (PFS) and overall survival (OS). Final PFS testing was completed at the second interim analysis (IA2; data cutoff, December 3, 2021); OS was tested at final analysis (FA; data cutoff, January 17, 2023). Efficacy boundaries were one-sided P = .0117 for PFS and OS.
RESULTS
Four hundred ninety-two patients were randomly assigned to pembrolizumab plus chemotherapy (n = 245) or placebo plus chemotherapy (n = 247). At IA2, the median PFS was 5.6 months for pembrolizumab plus chemotherapy versus 5.5 months for placebo plus chemotherapy (hazard ratio [HR], 0.80 [95% CI, 0.65 to 0.97]; P = .0122). At FA, the median OS was 15.9 versus 14.7 months, respectively (HR, 0.84 [95% CI, 0.69 to 1.02]; P = .0362). Grade ≥3 treatment-related adverse events occurred in 43.7% of pembrolizumab plus chemotherapy recipients versus 38.6% of placebo plus chemotherapy recipients.
CONCLUSION
Addition of pembrolizumab to chemotherapy in patients with TKI-resistant, EGFR-mutant, metastatic nonsquamous NSCLC did not significantly prolong PFS or OS versus placebo plus chemotherapy in KEYNOTE-789.
INTRODUCTION
Immune checkpoint inhibitors (ICIs), particularly anti‒PD-1 and anti‒PD-L1 antibodies, have greatly improved outcomes in advanced or metastatic non–small cell lung cancer (NSCLC).1 However, patients with mutations in epidermal growth factor receptor (EGFR) have been excluded from many of the anti‒PD-(L)1-based clinical trials because of initial data suggesting less efficacy of anti‒PD-(L)1 monotherapy in this population and the availability of tyrosine kinase inhibitors (TKIs).2-7 Standard first-line treatment for EGFR-mutant NSCLC is third-generation TKIs2,7; however, most patients develop treatment resistance and experience disease progression (PD).8,9 Subsequent treatment options have limited clinical benefit.4,6,10,11 These options include local therapy, osimertinib for T790M-positive tumors previously untreated with an approved third-generation TKI or previously treated with first- or second-generation TKIs, continuation of the same TKI therapy beyond progression, platinum-based chemotherapy, and atezolizumab (anti‒PD-L1) plus bevacizumab (anti–vascular endothelial growth factor [VEGF] antibody) and chemotherapy.2,7 In addition, single-agent ICI may be considered on failure of TKIs and chemotherapy.2,7
CONTEXT
Key Objective
The phase III KEYNOTE-789 study examined whether patients with metastatic nonsquamous non–small cell lung cancer (NSCLC) with an epidermal growth factor receptor (EGFR) mutation previously treated with tyrosine kinase inhibitor (TKI) would benefit from treatment with pembrolizumab plus pemetrexed and platinum chemotherapy versus chemotherapy alone.
Knowledge Generated
Pembrolizumab plus pemetrexed-platinum did not significantly prolong progression-free survival or overall survival versus pemetrexed-platinum alone. There were no unexpected toxicities or detrimental impact on quality of life.
Relevance (T.E. Stinchcombe)
For patients with EGFR mutant NSCLC with progressive disease after an EGFR TKI, the combination of platinum-pemetrexed and pembrolizumab should not be used as a subsequent therapy.*
*Relevance section written by JCO Associate Editor Thomas E. Stinchcombe, MD.
Pembrolizumab, an anti‒PD-1 monoclonal antibody, as monotherapy12,13 and in combination with chemotherapy,14,15 has significantly improved survival outcomes in previously untreated, advanced, or metastatic NSCLC without EGFR (or ALK) alterations versus chemotherapy alone. Specifically, in the phase III KEYNOTE-189 study, pembrolizumab plus chemotherapy significantly improved overall survival (OS; hazard ratio [HR], 0.49; 95% CI, 0.38 to 0.64; P < .001) and progression-free survival (PFS; HR, 0.52 [95% CI, 0.43 to 0.64]; P < .001) in previously untreated metastatic nonsquamous NSCLC, irrespective of PD-L1 tumor proportion score (TPS).14 In light of these data, there was an interest in assessing pembrolizumab plus chemotherapy further in EGFR-mutant NSCLC since patients with these mutations were not eligible for inclusion in KEYNOTE-189.
In the phase III KEYNOTE-789 study, we assessed the efficacy and safety of pemetrexed plus platinum chemotherapy with or without pembrolizumab in TKI-resistant, EGFR-mutant, metastatic nonsquamous NSCLC, irrespective of PD-L1 TPS (ClinicalTrials.gov: NCT03515837).
METHODS
Study Design and Patients
This randomized, double-blind, phase III study was conducted globally across 139 sites in accordance with Good Clinical Practice guidelines. The study protocol (online only) and amendments were approved by the appropriate ethics body at each site. All patients provided written informed consent before participation. An external data monitoring committee assessed the overall risk-benefit ratio of the trial, safety at periodic intervals, and efficacy at one of the prespecified interim analyses (IAs).
Key eligibility criteria included patients 18 years and older with histologically or cytologically confirmed stage IV nonsquamous NSCLC (per American Joint Committee on Cancer, 8th edition), an Eastern Cooperative Oncology Group performance status of 0 or 1, measurable disease per RECIST version 1.1, tumor tissue sample for assessment, adequate organ function, locally assessed and documented DEL19 or L858R EGFR mutation, and investigator-determined radiographic PD per RECIST version 1.1 after EGFR-TKI treatment. Patients whose disease progressed on first-line osimertinib were eligible regardless of T790M mutation status, whereas patients whose disease progressed on erlotinib, afatinib, or gefitinib must have also progressed on osimertinib if T790M mutation was detected. Patients with previously treated, radiologically stable brain metastases were eligible.
Treatment
Patients were randomly assigned 1:1 to pembrolizumab 200 mg or saline placebo once every 3 weeks for up to 35 cycles (approximately 2 years). All patients also received pemetrexed 500 mg/m2 and investigator's choice of cisplatin 75 mg/m2 or carboplatin area under the curve 5 mg/mL/min once every 3 weeks for four cycles followed by pemetrexed 500 mg/m2 maintenance therapy. Treatment continued until the maximum number of cycles (except pemetrexed as maintenance) or until PD as confirmed by blinded independent central review (BICR) per RECIST version 1.1, unacceptable toxicity, pregnancy, noncompliance, investigator's decision, or patient withdrawal. Patients assigned to placebo–chemotherapy who experienced PD could cross over to receive pembrolizumab monotherapy on study for a total of 35 cycles if other eligibility criteria were met. Random allocation was performed via an integrated interactive voice- and web-response system and stratified by PD-L1 TPS (<50% v ≥ 50%), previous osimertinib (yes v no), and region (East Asia v non‒East Asia).
Assessments
PD-L1 expression was centrally evaluated in archival/newly obtained formalin-fixed, paraffin-embedded tumor samples collected from nonirradiated lesions using PD-L1 IHC 22C3 pharmDx (Agilent Technologies, Carpinteria, CA), and categorized by TPS (percentage of tumor cells with membranous PD-L1 staining). Tumor imaging was performed at baseline, 6 weeks after random assignment, every 9 weeks through 51 weeks, and every 12 weeks thereafter (Appendix 1, online only). In patients who received crossover treatment, tumor imaging was performed within 4 weeks before receiving pembrolizumab and every 12 weeks thereafter. Tumor response was assessed per RECIST version 1.1 by BICR for determination of study end points and eligibility and according to iRECIST16 by investigator for treatment decisions after PD. Adverse events (AEs) were recorded from random assignment through 30 days (90 days for serious AEs) after last dose of study treatment. AEs were assessed and graded by the investigator according to the National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.0. Survival was assessed every 12 weeks during follow-up. Patient-reported outcomes (PROs) were evaluated using the European Organisation for Research and Treatment of Cancer (EORTC) Quality-of-Life Questionnaire‒Core 30 (QLQ-C30), the 13-item EORTC QLQ‒Lung Cancer Module (LC13), and the EuroQoL 5-dimension, 5-level questionnaire.
End Points
The dual primary end points were PFS (time from random assignment to first of PD or death) and OS (time from random assignment to death from any cause). Secondary end points included objective response rate (ORR; proportion of patients with complete response [CR] or partial response [PR]), duration of response (DOR; time from evidence of response to first of PD or death in patients with CR or PR), PROs, and safety. The protocol-specified secondary PRO end points included mean score change from baseline to week 18 in the global health status (GHS)/quality of life (QoL) scale (GHS item 29; QoL item 30) and the time to true deterioration (TTD; time from baseline to first onset of ≥10 points of deterioration from baseline, with confirmed second adjacent deterioration of ≥10 points from baseline in any of the three symptoms) in the composite end point of cough (QLQ-LC13 item 1), chest pain (QLQ-LC13 item 10), or dyspnea (QLQ-C30 item 8).
Statistical Analysis
Efficacy was assessed in the intention-to-treat population (all randomly assigned patients), safety in all randomly assigned patients who received ≥1 dose of study treatment, and PROs in all patients with ≥1 PRO assessment available and ≥1 dose of study treatment. PFS, OS, DOR, and TTD were assessed using the Kaplan-Meier method; censoring rules are summarized in the Appendix (Table A1). Between-group differences in OS, PFS, and TTD were assessed using the stratified log-rank test. All P values are one-sided. A stratified Cox proportional hazards model with Efron's method of tie handling was used to estimate the magnitude of treatment difference (HR) between treatment groups. Treatment difference in ORR was assessed using a stratified Miettinen and Nurminen method. Change in least squares mean in the GHS/QoL score from baseline to week 18 was assessed using a constrained longitudinal data analysis model, with GHS/QOL score as the response variable and treatment by study visit interaction and stratification factors as covariates. All stratified analyses were performed using the random assignment stratification factors.
The protocol specified three IAs and a final analysis (FA). Second interim analysis (IA2) was the prespecified final PFS analysis. IA3 became the FA because the observed number of OS events at IA3 was close to the target number for FA. The graphical method of Maurer and Bretz was used to control the family-wise type I error rate at 0.025 (one-sided) for hypothesis testing of PFS, OS, and ORR, with preallocated α at .0125, .0125, and 0, respectively (Appendix Fig A1). With 492 patients with ≥24 months of enrollment, the study had approximately 91% power to detect an HR of 0.7 for PFS and approximately 86% power to detect an HR of 0.72 for OS at FA, assuming 423 deaths and 3 IAs, with overall α = .0125 (1-sided). IA1 was planned for approximately 6 months after the last patient was randomly assigned and on the basis of a data cutoff of February 3, 2021. IA2 was planned for approximately 16 months after the last patient was randomly assigned and approximately 414 PFS events were observed. IA2 was based on a data cutoff of December 3, 2021. IA3 (became FA) was planned approximately 29 months after the last patient was randomly assigned and on the basis of a data cutoff of January 17, 2023. Only OS was tested formally at FA. ORR was only to be tested at IA when either PFS or OS was significant. All superiority boundaries were calculated using the Lan-DeMets O'Brien-Fleming spending function. On the basis of the actual number of events observed, the efficacy boundaries were P = .0117 for PFS (at IA2) and OS (at FA) per the protocol-specified minimum spending approach.
RESULTS
Patients
Between July 23, 2018, and August 30, 2020, 492 patients were randomly allocated to receive pembrolizumab–chemotherapy (n = 245) or placebo–chemotherapy (n = 247; Fig 1). Demographics and baseline disease characteristics were generally balanced between groups (Table 1). The median age was 63 years, 66.1% of patients were never smokers, 48.2% received either first-line or second-line osimertinib, and 20.9% had tumors with PD-L1 TPS ≥50%.
TABLE 1.
Characteristic | Pembrolizumab Plus Pemetrexed-Platinum (n = 245) | Placebo Plus Pemetrexed-Platinum (n = 247) |
---|---|---|
Age, years, median (range) | 62.0 (34‒87) | 64.0 (34‒84) |
Sex | ||
Male | 93 (38.0) | 96 (38.9) |
Female | 152 (62.0) | 151 (61.1) |
ECOG performance status | ||
0 | 71 (29.0) | 92 (37.2) |
1 | 174 (71.0) | 155 (62.8) |
Chemotherapy received | ||
Cisplatin/pemetrexed | 37 (15.1) | 43 (17.4) |
Carboplatin/pemetrexed | 208 (84.9) | 203 (82.2) |
Missing | 0 | 1 (0.4) |
Region | ||
East Asia | 150 (61.2) | 150 (60.7) |
Non-East Asia | 95 (38.8) | 97 (39.3) |
Smoking status | ||
Former or current | 84 (34.3) | 83 (33.6) |
Never | 161 (65.7) | 164 (66.4) |
Histology | ||
Adenocarcinoma | 239 (97.6) | 243 (98.4) |
NSCLC not otherwise specified | 5 (2.0) | 2 (0.8) |
Other | 1 (0.4) | 2 (0.8) |
Brain metastases at baseline | 51 (20.8) | 47 (19.0) |
History of brain metastases | 69 (28.2) | 69 (27.9) |
Liver metastases at baseline | 54 (22.0) | 40 (16.2) |
PD-L1 status | ||
TPS ≥50% | 52 (21.2) | 51 (20.6) |
TPS 1%-49% | 54 (22.0) | 72 (29.1) |
TPS <1% | 127 (51.8) | 113 (45.7) |
Not evaluable | 12 (4.9) | 11 (4.5) |
Previous use of TKI | ||
Any first- or second-generation TKI | 128 (52.2) | 126 (51.0) |
First-line osimertinib | 28 (11.4) | 33 (13.4) |
Second-line osimertinib | 88 (35.9) | 88 (35.6) |
Other | 1 (0.4) | 0 |
Previous therapy | ||
Neoadjuvant therapy | 1 (0.4) | 4 (1.6) |
Adjuvant therapy | 13 (5.3) | 12 (4.9) |
Radiation | 81 (33.1) | 92 (37.2) |
T790M mutation | ||
Positive | 95 (38.8) | 87 (35.2) |
Negative | 129 (52.7) | 140 (56.7) |
Not done | 21 (8.6) | 20 (8.1) |
EGFR-activating mutation | ||
L858R | 103 (42.0) | 102 (41.3) |
DEL19 | 139 (56.7) | 142 (57.5) |
L858R and DEL19 | 2 (0.8) | 2 (0.8) |
Other | 1 (0.4) | 1 (0.4) |
NOTE. Data are No. (%) unless noted otherwise.
Abbreviations: ECOG, Eastern Cooperative Oncology Group; EGFR, epidermal growth factor receptor; NSCLC, non–small cell lung cancer; TKI, tyrosine kinase inhibitor; TPS, tumor proportion score.
At least one dose of study treatment was received by 245 patients in the pembrolizumab–chemotherapy group and 246 patients in the placebo–chemotherapy group. In the pembrolizumab–chemotherapy group, 240 patients (98.0%) discontinued all treatment, three (1.2%) completed treatment, and two (0.8%) remained on treatment (Fig 1). Eighteen patients (7.3%) began subsequent anti‒PD-(L)1 therapy, and 96 (39.2%) were retreated with TKIs. In the placebo–chemotherapy group, 243 patients (98.8%) had discontinued, two (0.8%) completed, and one (0.4%) remained on treatment (Fig 1). At data cutoff, 50 patients (20.2%) from the placebo–chemotherapy group had crossed over to receive pembrolizumab monotherapy on study (Appendix Table A2) and an additional 38 (15.4%) received anti‒PD-(L)1 as subsequent therapy outside the study, for an effective crossover rate of 35.6%. In addition, 101 patients (40.9%) in the placebo–chemotherapy group received subsequent TKIs.
Efficacy
At IA2, with a median time from random assignment to data cutoff of 28.6 (16.0 to 40.4) months, 412 events of PD or death had occurred. The median (95% CI) PFS was 5.6 (5.5 to 5.8) months in the pembrolizumab–chemotherapy group versus 5.5 (5.4 to 5.6) months with placebo–chemotherapy. The HR for PFS was 0.80 (95% CI, 0.65 to 0.97; P = .0122; Fig 2A). PFS outcomes were similar in most subgroups, including by PD-L1 TPS (Figs 2B, 3A, and 3B, Appendix Figs A2A and A2B).
At FA, the median (range) time from random assignment to data cutoff was 42.0 (29.5 to 53.9) months and there were 438 deaths. The median OS (95% CI) was 15.9 (13.7 to 18.8) months for pembrolizumab–chemotherapy and 14.7 (12.7 to 17.1) months for placebo–chemotherapy. The HR for OS was 0.84 (95% CI, 0.69 to 1.02; P = .0362; Fig 2C). OS outcomes were similar across most subgroups analyzed (Fig 2D, Appendix Figs A2C and A2D). There was a trend toward improved OS in patients with PD-L1 TPS ≥1% for pembrolizumab–chemotherapy versus placebo–chemotherapy compared with PD-L1 TPS <1% (Figs 3C and 3D).
At FA, the ORR (95% CI) was 29.0% (23.4 to 35.1) for pembrolizumab–chemotherapy and 27.1% (21.7 to 33.1) for placebo–chemotherapy (Table 2).
TABLE 2.
Response | Pembrolizumab Plus Pemetrexed-Platinum (n = 245) | Placebo Plus Pemetrexed-Platinum (n = 247) |
---|---|---|
ORR (95% CI), % | 29.0 (23.4 to 35.1) | 27.1 (21.7 to 33.1) |
Best overall response, No. (%) | ||
CR | 5 (2.0) | 3 (1.2) |
PR | 66 (26.9) | 64 (25.9) |
SDa | 121 (49.4) | 117 (47.4) |
PD | 37 (15.1) | 52 (21.1) |
Not evaluableb | 8 (3.3) | 5 (2.0) |
No assessmentc | 8 (3.3) | 6 (2.4) |
DOR,d median (range), months | 6.3 (2.3-40.8+)e | 5.6 (1.8+-40.6+)e |
DOR ≥9 months,d No. (%) | 23 (34.0) | 13 (22.9) |
Time to response, median (range), months | 1.5 (1.2-9.7) | 1.5 (1.2-25.3) |
Abbreviations: CR, complete response; DOR, duration of response; ITT, intention to treat; ORR, objective response rate; PD, progressive disease; PR, partial response; SD, stable disease.
Stable disease includes stable disease, non-CR/non-PD, and no evidence of disease (no lesions identified at baseline or postbaseline assessments).
Postbaseline assessment(s) available but not evaluable or CR/PR/SD <6 weeks from random assignment.
No postbaseline assessment available for response evaluation.
On the basis of Kaplan-Meier estimate.
The + indicates no progressive disease by the time of last disease assessment.
In patients who crossed over from placebo–chemotherapy to pembrolizumab monotherapy on study (n = 50), the median PFS (95% CI) from the time of beginning pembrolizumab monotherapy to first documented PD or death was 7.3 (4.8 to 8.5) months.
Safety
The median (range) number of treatment cycles was 8 (1‒62) for pembrolizumab–chemotherapy and 8 (1‒63) for placebo–chemotherapy. The median duration of treatment was 5.3 (0.03‒43.11) months and 5.0 (0.03‒42.88) months, respectively. AEs of any grade occurred in 239 of 245 treated patients (97.6%) in the pembrolizumab–chemotherapy group and 241 of 246 patients (98.0%) in the placebo–chemotherapy group (Table 3). Treatment-related AEs occurred in 220 patients (89.8%) and 212 (86.2%), respectively. These were grade ≥3 in 107 (43.7%) and 95 (38.6%) patients, respectively. There was one fatal treatment-related AE due to myocarditis in the pembrolizumab–chemotherapy group and two in the placebo–chemotherapy group due to bone marrow failure and general physical health deterioration.
TABLE 3.
Event | Pembrolizumab Plus Pemetrexed-Platinum (n = 245), No. (%) | Placebo Plus Pemetrexed-Platinum (n = 246), No. (%) | ||
---|---|---|---|---|
Any AE (all-cause) | 239 (97.6) | 241 (98.0) | ||
Grade 3 to 5 | 137 (55.9) | 143 (58.1) | ||
Led to deatha | 5 (2.0) | 12 (4.9) | ||
Treatment-related | 220 (89.8) | 212 (86.2) | ||
Grade 3 to 5 | 107 (43.7) | 95 (38.6) | ||
Led to discontinuation of any treatment component | 40 (16.3) | 29 (11.8) | ||
Led to discontinuation of pembrolizumab or placebo | 24 (9.8) | 11 (4.5) | ||
Led to discontinuation of any chemotherapy | 31 (12.7) | 29 (11.8) | ||
Led to discontinuation of all treatment components | 7 (2.9) | 5 (2.0) | ||
Treatment-related AEs occurring in ≥10% of patients in either treatment group | Any Grade | Grade 3‒5 | Any Grade | Grade 3‒5 |
Anemia | 94 (38.4) | 31 (12.7) | 99 (40.2) | 23 (9.3) |
Decreased neutrophil count | 92 (37.6) | 56 (22.9) | 111 (45.1) | 52 (21.1) |
Nausea | 85 (34.7) | 5 (2.0) | 102 (41.5) | 8 (3.3) |
Decreased white blood cell count | 74 (30.2) | 26 (10.6) | 86 (35.0) | 22 (8.9) |
Fatigue | 57 (23.3) | 8 (3.3) | 44 (17.9) | 5 (2.0) |
Increased alanine aminotransferase | 55 (22.4) | 10 (4.1) | 51 (20.7) | 6 (2.4) |
Increased aspartate aminotransferase | 54 (22.0) | 4 (1.6) | 50 (20.3) | 2 (0.8) |
Decreased platelet count | 54 (22.0) | 21 (8.6) | 49 (19.9) | 13 (5.3) |
Decreased appetite | 53 (21.6) | 7 (2.9) | 49 (19.9) | 1 (0.4) |
Constipation | 47 (19.2) | 2 (0.8) | 41 (16.7) | 1 (0.4) |
Vomiting | 36 (14.7) | 2 (0.8) | 34 (13.8) | 6 (2.4) |
Asthenia | 25 (10.2) | 5 (2.0) | 20 (8.1) | 1 (0.4) |
Immune-mediated AEs and infusion reactions | 49 (20.0) | 11 (4.5) | 20 (8.1) | 5 (2.0) |
Hypothyroidism | 14 (5.7) | 0 | 6 (2.4) | 0 |
Hyperthyroidism | 13 (5.3) | 0 | 3 (1.2) | 0 |
Infusion reactions | 5 (2.0) | 0 | 2 (0.8) | 0 |
Pneumonitis | 5 (2.0) | 2 (0.8) | 4 (1.6) | 2 (0.8) |
Adrenal insufficiency | 2 (0.8) | 0 | 0 | 0 |
Hepatitis | 2 (0.8) | 2 (0.8) | 1 (0.4) | 1 (0.4) |
Myositis | 2 (0.8) | 2 (0.8) | 1 (0.4) | 0 |
Nephritis | 2 (0.8) | 1 (0.4) | 1 (0.4) | 1 (0.4) |
Severe skin reactions | 2 (0.8) | 2 (0.8) | 0 | 0 |
Thyroiditis | 2 (0.8) | 0 | 3 (1.2) | 0 |
Colitis | 1 (0.4) | 1 (0.4) | 1 (0.4) | 1 (0.4) |
Hypophysitis | 1 (0.4) | 0 | 0 | 0 |
Myelitis | 1 (0.4) | 0 | 0 | 0 |
Myocarditis | 1 (0.4) | 1 (0.4) | 0 | 0 |
Type 1 diabetes mellitus | 1 (0.4) | 1 (0.4) | 0 | 0 |
Abbreviation: AE, adverse event.
One patient in the pembrolizumab plus pemetrexed-platinum group died because of a treatment-related AE of myocarditis; two patients in the placebo plus pemetrexed-platinum group died because of treatment-related AEs of bone marrow failure (n = 1) and general physical health deterioration (n = 1).
Immune-mediated AEs and infusion reactions occurred in 49 patients (20.0%) who received pembrolizumab–chemotherapy and 20 (8.1%) in the placebo–chemotherapy group. These were grade ≥3 in 11 (4.5%) and five (2.0%) patients, respectively. The only fatal immune-mediated event was myocarditis (0.4%; noted above) in the pembrolizumab–chemotherapy group.
PROs
The PRO analysis population comprised 473 patients (pembrolizumab–chemotherapy, n = 237; placebo–chemotherapy, n = 236) who completed ≥1 QLQ-C30 and QLQ-LC13 assessment. At baseline, the completion and compliance rates were >97.0% for each treatment group and >71.2% at week 18 (Appendix Table A3).
At week 18, the least squares mean (95% CI) change from baseline in QLQ-C30 GHS/QoL scores was stable in both treatment groups (Appendix Table A4); no clinically meaningful between-group differences were observed (least squares mean difference 1.59 points; 95% CI, –1.93 to 5.10). The median (95% CI) TTD for the composite end point was not reached (10.05 months to not reached) for pembrolizumab–chemotherapy and 17.97 months (6.67 months to not reached) for placebo–chemotherapy (HR, 0.93 [95% CI, 0.68 to 1.27]; Fig 4).
DISCUSSION
In this study of TKI-resistant, EGFR-mutant, metastatic nonsquamous NSCLC, the addition of pembrolizumab to pemetrexed and platinum chemotherapy did not significantly prolong PFS and OS versus placebo plus chemotherapy per the prespecified statistical analysis plan. No new safety signals were identified in either treatment group. Pembrolizumab–chemotherapy demonstrated no clinically meaningful detriment in overall QoL and comparable TTD in disease-related symptoms versus placebo–chemotherapy. Among 50 patients who received pembrolizumab in the on-study crossover, 40% had PD-L1 TPS <1%. There was evidence of antitumor activity in these patients; however, no meaningful conclusions can be drawn because of small sample size and the non-prespecified nature of this analysis.
PFS and OS outcomes were similar in most subgroups assessed. No specific differences were observed in outcomes by choice of platinum chemotherapy (carboplatin or cisplatin), similar to other studies examining immunotherapy plus chemotherapy.14,17 There was a trend toward longer PFS and OS with pembrolizumab–chemotherapy versus placebo–chemotherapy in patients without brain metastasis at baseline; albeit, a similar trend was not observed in patients with brain metastasis at baseline, which represented a small subgroup with wide CIs. Trends in PD-L1 TPS subgroups also differed, with outcomes in patients with PD-L1 TPS ≥50% being similar between treatment groups, but a trend for improved OS with pembrolizumab–chemotherapy in patients with PD-L1 TPS ≥1% versus <1%. However, a similar trend was not observed for PFS. Importantly, these subgroup analyses were not adjusted for multiplicity, and no definitive conclusions can be drawn from these observations. Similar to these findings, previous studies of ICIs in EGFR-mutant NSCLC have shown a variable relationship between PD-L1 expression and outcomes to therapy, suggesting that PD-L1 remains an inconclusive biomarker for response in this setting.6,18-21 Overall, EGFR-mutant tumors have been documented to have immunosuppressive or cold microenvironments and lower tumor mutational burden compared with tumors without EGFR mutations and correlate with inferior response to anti–PD-(L)1 therapy.11,18
Results presented here follow those from phase II studies of pembrolizumab plus chemotherapy in TKI-resistant advanced EGFR-mutant NSCLC where the ORR ranged from 42% to 58%.22,23 However, these benefits were not realized in our larger and more robust study. Similarly, in CheckMate 722, a prospective phase III study of TKI-resistant, EGFR-mutant, metastatic NSCLC, there was no significant improvement in PFS (primary end point) with nivolumab (anti–PD-1) plus chemotherapy versus chemotherapy alone (HR, 0.75 [95% CI, 0.56 to 1.00]).24 However, the study did not reach the required number of PFS events and was therefore underpowered.24
By contrast, the phase III ORIENT-31 study showed significantly improved PFS outcomes (primary end point) in TKI-resistant, EGFR-mutant advanced or metastatic NSCLC, with sintilimab (anti‒PD-1) plus chemotherapy versus chemotherapy alone (P = .016), but without similar benefit in OS.25 Comparatively greater benefit was observed in a third treatment arm that added IBI305 (anti-VEGF) to sintilimab plus chemotherapy versus chemotherapy alone (P < .0001) although no direct comparison between all three study arms has been reported.26 Similarly, the ATTLAS study showed significant PFS benefit (primary end point) with atezolizumab plus bevacizumab and chemotherapy versus chemotherapy alone (P = .004) in patients with EGFR- or ALK-mutant metastatic NSCLC, but without OS benefit.21 These results built on a subgroup analysis (n = 124) of EGFR-mutant NSCLC from the phase III IMpower150 study of atezolizumab plus bevacizumab and chemotherapy, which showed a trend toward improved OS versus bevacizumab plus chemotherapy alone.10 Notably, no OS or PFS benefit was observed with atezolizumab plus chemotherapy (without bevacizumab) versus bevacizumab plus chemotherapy.10 While these studies suggest an added benefit of anti–VEGF therapy when combined with anti–PD-(L)1 therapy and chemotherapy, there were no statistically significant improvements in OS outcomes. Differences in outcomes between KEYNOTE-789 and these studies may be explained by several factors. First, moderate clinical benefit from anti-VEGF therapy has been reported for EGFR-mutant NSCLC, albeit with some increased toxicity.27-29 Other factors include different study demographics (ie, ORIENT-31 was conducted in China, and ATTLAS in South Korea) and previous osimertinib treatment. In CheckMate 722, for example, 23% of patients received previous osimertinib versus 48% in KEYNOTE-789.24 Similarly, 11% of patients received previous third-generation TKIs (including osimertinib) in ORIENT-3125 and only 10 patients received previous osimertinib in IMpower150.10 Because third-generation TKIs are the standard of care for these patients, the results from KEYNOTE-789 may more accurately reflect the real-world situation. In addition, treatment with TKIs can lead to a range of resistance mechanisms, changes in the tumor microenvironment to become more immunosuppressive, and decreased PD-L1 mRNA expression and PD-L1 degradation, which could decrease the response to anti–PD-(L)1-based therapies.30-32 There were no new safety signals with pembrolizumab–chemotherapy in KEYNOTE-789. The incidence of grade 3 to 5 treatment-related AEs was similar between treatment groups, as was the proportion of patients who discontinued treatment because of AEs. The safety profile of pembrolizumab–chemotherapy in KEYNOTE-789 was also comparable with that observed in patients with previously untreated, metastatic NSCLC without EGFR or ALK alterations.14,15
In conclusion, pembrolizumab plus pemetrexed and platinum chemotherapy did not significantly prolong PFS and OS versus chemotherapy alone in patients with EGFR-mutant, TKI-resistant metastatic nonsquamous NSCLC, per the prespecified statistical analysis plan. There were no unexpected toxicities, and no detrimental impact in QoL was associated with pembrolizumab–chemotherapy compared with chemotherapy alone. Evidence to date suggests that anti‒PD-(L)1 plus chemotherapy may not provide sufficient clinical benefit compared with chemotherapy alone, and a better understanding of potential biomarkers may be required to identify patients who will experience the full benefit of anti‒PD-(L)1 therapy in TKI-resistant, EGFR-mutant, metastatic nonsquamous NSCLC.
ACKNOWLEDGMENT
We thank the patients and their families and caregivers for participating in this study, along with all investigators and site personnel. Eli Lilly provided pemetrexed but had no additional role in trial conduct. Medical writing assistance was provided by Sonia Mohinta, PhD, and Kathleen Estes, PhD, of ICON plc (Blue Bell, PA, USA). This assistance was funded by MSD. The list of investigators is provided in Appendix Table A5.
APPENDIX Description of Brain Metastasis Imaging Requirements in KEYNOTE-789
Brain magnetic resonance imaging was required for all patients at screening, unless medically contraindicated; computed tomography with contrast was allowed in those instances. Ideally, for a given patient, the same type of scan, same scanner, and the use of contrast were recommended throughout the study to optimize the reproducibility and accuracy of the assessment of existing and new tumor burden. All images were submitted to the central imaging vendor, and radiographic progression per RECIST version 1.1 was confirmed by the central imaging vendor. Imaging assessments were performed at 6 weeks from the date of random assignment, then every 9 weeks until week 51 (or more frequently if clinically indicated), and every 12 weeks thereafter. For imaging throughout the study, on-study brain or bone imaging was performed if clinically indicated or to confirm complete response (if other lesions indicated complete response and brain or bone lesions existed at baseline).
TABLE A1.
Situation | Censoring Rule |
---|---|
Progression-free survival | |
PD or death documented after ≤1 missed disease assessment, and before any new anticancer therapy, if any | Progressed at the date of documented PD or death |
PD or death documented immediately after ≥2 consecutive missed disease assessments or after new anticancer therapy, if any | Censored at last disease assessment before the earlier date of ≥2 consecutive missed disease assessments and new anticancer therapy, if any |
No PD, no death; new anticancer treatment is not initiated | Censored at last disease assessment |
No PD, no death; new anticancer treatment is initiated | Censored at last disease assessment before new anticancer treatment |
Overall survival | |
No death | Censored at the last date patient was known to be alive |
Duration of responsea | |
No progression nor death, no new anticancer treatment initiated | Censored at last adequate disease assessment |
No progression nor death, new anticancer treatment initiated | Censored at last adequate disease assessment before new anticancer therapy |
Death or progression immediately after ≥2 consecutive missed disease assessments or after new anticancer therapy, if any | Censored at the earlier date of last adequate disease assessment before ≥2 missed adequate disease assessments and new anticancer therapy, if any |
Death or progression after ≤1 missed disease assessment and before the new anticancer therapy, if any | End of response at the date of PD or death |
Time to deterioration | |
Deterioration confirmed | Event observed at the time of assessment (first deterioration) |
Ongoing or discontinued from the study without confirmed deterioration | Right censored at the time of last assessment |
No baseline assessments | Right censored at treatment start date |
Abbreviation: PD, progressive disease.
For duration of response, a missed disease assessment includes any assessment that is not obtained or is considered inadequate for evaluation of response.
TABLE A2.
Characteristic | Patients Who Crossed Over to Pembrolizumab From Placebo (n = 50) |
---|---|
Age, years, median (range) | 65.0 (45-79) |
Sex | |
Male | 18 (36.0) |
Female | 32 (64.0) |
ECOG performance status | |
0 | 17 (34.0) |
1 | 33 (66.0) |
Chemotherapy received | |
Cisplatin/pemetrexed | 11 (22.0) |
Carboplatin/pemetrexed | 39 (78.0) |
Region | |
East Asia | 38 (76.0) |
Non-East Asia | 12 (24.0) |
Smoking status | |
Former or current | 15 (30.0) |
Never | 35 (70.0) |
Adenocarcinoma histology | 50 (100.0) |
Brain metastases at baseline | 9 (18.0) |
History of brain metastases | 11 (22.0) |
Liver metastases at baseline | 11 (22.0) |
PD-L1 status | |
TPS ≥50% | 13 (26.0) |
TPS 1%-49% | 15 (30.0) |
TPS <1% | 20 (40.0) |
Not evaluable | 2 (4.0) |
Previous use of TKI | |
Any first or second generation TKI | 28 (56.0) |
First-line osimertinib | 4 (8.0) |
Second-line osimertinib | 18 (36.0) |
Previous therapy | |
Neoadjuvant therapy | 1 (2.0) |
Adjuvant therapy | 2 (4.0) |
Radiation | 14 (28.0) |
T790M mutation | |
Positive | 18 (36.0) |
Negative | 29 (58.0) |
Not done | 3 (6.0) |
EGFR-activating mutation | |
L858R | 17 (34.0) |
DEL19 | 33 (66.0) |
NOTE. Data are No. (%) unless noted otherwise.
Abbreviations: ECOG, Eastern Cooperative Oncology Group; EGFR, epidermal growth factor receptor; NSCLC, non–small cell lung cancer; TKI, tyrosine kinase inhibitor; TPS, tumor proportion score.
TABLE A3.
Treatment Visit | QLQ-C30 | QLQ-LC13 | ||
---|---|---|---|---|
Pembrolizumab Plus Pemetrexed-Platinum (n = 241) | Placebo Plus Pemetrexed-Platinum (n = 243) | Pembrolizumab Plus Pemetrexed-Platinum (n = 241) | Placebo Plus Pemetrexed-Platinum (n = 243) | |
Baseline | ||||
Completion | 237/241 (98.3) | 236/243 (97.1) | 237/241 (98.3) | 236/243 (97.1) |
Compliance | 237/241 (98.3) | 236/243 (97.1) | 237/241 (98.3) | 236/243 (97.1) |
Week 3 | ||||
Completion | 209/241 (86.7) | 219/243 (90.1) | 208/241 (86.3) | 219/243 (90.1) |
Compliance | 209/223 (93.7) | 219/228 (96.1) | 208/223 (93.3) | 219/228 (96.1) |
Week 6 | ||||
Completion | 198/241 (82.2) | 205/243 (84.4) | 196/241 (81.3) | 205/243 (84.4) |
Compliance | 198/216 (91.7) | 205/220 (93.2) | 196/216 (90.7) | 205/220 (93.2) |
Week 9 | ||||
Completion | 181/241 (75.1) | 195/243 (80.2) | 182/241 (75.5) | 194/243 (79.8) |
Compliance | 181/196 (92.3) | 195/213 (91.5) | 182/196 (92.9) | 194/213 (91.1) |
Week 12 | ||||
Completion | 175/241 (72.6) | 177/243 (72.8) | 174/241 (72.2) | 177/243 (72.8) |
Compliance | 175/183 (95.6) | 177/191 (92.7) | 174/183 (95.1) | 177/191 (92.7) |
Week 15 | ||||
Completion | 165/241 (68.5) | 167/243 (68.7) | 164/241 (68.0) | 165/243 (67.9) |
Compliance | 165/177 (93.2) | 167/186 (89.8) | 164/177 (92.7) | 165/186 (88.7) |
Week 18 | ||||
Completion | 173/241 (71.8) | 173/243 (71.2) | 171/241 (71.0) | 173/243 (71.2) |
Compliance | 173/179 (96.6) | 173/178 (97.2) | 171/179 (95.5) | 173/178 (97.2) |
Week 27 | ||||
Completion | 127/241 (52.7) | 121/243 (49.8) | 127/241 (52.7) | 120/243 (49.4) |
Compliance | 127/147 (86.4) | 121/147 (82.3) | 127/147 (86.4) | 120/147 (81.6) |
Week 36 | ||||
Completion | 89/241 (36.9) | 77/243 (31.7) | 88/241 (36.5) | 77/243 (31.7) |
Compliance | 89/100 (89.0) | 77/93 (82.8) | 88/100 (88.0) | 77/93 (82.8) |
Week 45 | ||||
Completion | 61/241 (25.3) | 49/243 (20.2) | 61/241 (25.3) | 49/243 (20.2) |
Compliance | 61/74 (82.4) | 49/55 (89.1) | 61/74 (82.4) | 49/55 (89.1) |
Week 54 | ||||
Completion | 46/241 (19.1) | 34/243 (14.0) | 46/241 (19.1) | 33/243 (13.6) |
Compliance | 46/52 (88.5) | 34/40 (85.0) | 46/52 (88.5) | 33/40 (82.5) |
NOTE. Completion was defined as the proportion of patients who completed a PRO assessment among the total PRO analysis population. Compliance was defined as the proportion of patients who completed a PRO assessment among those expected to complete the instruments at each visit (excluding patients who discontinued study treatment).
Abbreviations: EORTC, European Organisation for Research and Treatment of Cancer; PRO, patient-reported outcome; QLQ-C30, Quality-of-Life Questionnaire‒Core 30; QLQ-LC13, 13-item EORTC QLQ‒Lung Cancer Module.
TABLE A4.
Time Point | Pembrolizumab Plus Pemetrexed-Platinum (n = 241) | Placebo Plus Pemetrexed-Platinum (n = 243) |
---|---|---|
Baseline, No.a | 237 | 236 |
Mean (SD) score | 65.65 (20.88) | 67.30 (19.65) |
Week 18, No.a | 172 | 173 |
Mean (SD) score | 68.65 (17.44) | 66.91 (18.12) |
Change from baseline, No.b | 241 | 243 |
LS mean score (95% CI) | –0.46 (–3.17 to 2.25) | –2.05 (–4.74 to 0.64) |
Between-group difference in LS mean (95% CI) | 1.59 (–1.93 to 5.10) |
Abbreviations: EORTC, European Organisation for Research and Treatment of Cancer; GHS/QoL, global health status/quality of life; QLQ-C30, Quality-of-Life Questionnaire‒Core 30.
No. represents the number of patients in each treatment group with nonmissing assessments at the specific time point.
No. represents the number of patients in the analysis population in each treatment group.
TABLE A5.
Country/Region | Principal Investigator |
---|---|
Australia | Michael Joseph Boyer |
Rina Hui/Mark Wong | |
Andrew Mant/Phillip Parente | |
Thomas John/Sagun Parakh | |
Brazil | Gilberto Castro, Jr |
Gustavo Werutsky | |
Sergio Jobim de Azevedo | |
Fabio Andre Franke | |
Joilda Batista De Almeida Rego | |
Pedro Rafael Martins De Marchi/Gustavo Dix Junqueira | |
Fernanda Maris Peria | |
Leandro Brust | |
Canada | Parneet Cheema |
Mark Doherty/Ambika Parmar/Ines B. Menjak | |
Natasha B. Leighl | |
Jason Agulnik | |
China | Shun Lu |
Zhigang Han | |
Jiuwei Cui | |
Li Zhang | |
Ying Cheng | |
Gongyan Chen | |
Helong Zhang | |
Yu Yao | |
Chengping Hu | |
Qiming Wang | |
Xin Zhang/Yong Zhang | |
Jianying Zhou | |
Kejing Ying | |
Yun Fan | |
Yan Wang | |
Ziping Wang | |
Jifeng Feng | |
YingYing Du | |
Lin Wu | |
Cheng Huang | |
Xiangdong Zhou | |
France | Maurice Perol |
Julien Domont | |
Corinne Lamour/Julien Dutilh | |
Youssef Oulkhouir | |
Virginie Westeel | |
Delphine Carmier | |
Bruno Coudert/Aurelie Lagrange | |
Dominique Spaeth | |
Stanislas Ropert | |
Germany | Daniel C. Christoph |
Jens Kern | |
Hans-Georg Kopp | |
Frank Griesinger | |
Rainer Wiewrodt | |
Martin Wermke | |
Claas Wesseler | |
Annette Mueller | |
Gunther Vogel | |
Hong Kong | Victor Lee |
Chung Man James Ho | |
Siu Hong Oscar Chan | |
Sing Hung Lo/Shi Feng Jonathan Nyaw | |
Yu Chung Jacky Li | |
Israel | Jair Bar |
Maya Gottfried | |
Julia Dudnik | |
Alona Zer/Mor Moskovitz/Mirjana Wollner | |
Ofer Rotem | |
Sivan Shamai | |
Noam Asna | |
Mhameed Kamel | |
Italy | Silvia Novello |
Francesco Di Costanzo/Laura Doni/Francesca Mazzoni | |
Francesco Ferrau | |
Filippo de Marinis | |
Giuseppe Tonini | |
Domenico Galetta | |
Francovito Piantedosi/Fabiana Vitiello | |
Japan | Keisuke Kirita/Kiyotaka Yoh |
Toshiaki Takahashi | |
Yuichiro Ohe | |
Yoshihiro Hattori | |
Isamu Okamoto | |
Takayasu Kurata/Hiroshige Yoshioka | |
Hideo Saka/Masahide Oki | |
Terufumi Kato | |
Hiroshi Tanaka | |
Toru Kumagai/Takako Inoue | |
Toyoaki Hida/Yoshitsugu Horio | |
Shunsuke Teraoka | |
Eiki Ichihara | |
Kazuma Kishi/Hisashi Takaya/Daiya Takai | |
Toshiyuki Kozuki | |
Kazuo Kasahara/Yuichi Tambo | |
Yukio Hosomi | |
Masashi Kondo | |
Masao Ichiki/Hiroaki Takeoka | |
Mexico | Emmanuel de la Mora Jimenez |
Carlos Alberto Hernandez | |
Jeronimo Rafael Rodriguez Cid | |
Oscar Gerardo Arrieta Rodriguez | |
South Korea | Ji-Youn Han |
Young Joo Min | |
Dong-Wan Kim | |
Keunchil Park/Se-Hoon Lee | |
Ki Hyeong Lee | |
Jong-Seok Lee | |
Jin Hyoung Kang | |
Dae Ho Lee | |
Eun Kyung Cho | |
Spain | Enric Carcereny |
Pilar Garrido Lopez | |
Margarita Majem Tarruella | |
Luis Paz-Ares Rodriguez | |
David Vicente Baz | |
Enriqueta Felip Font | |
Manuel Cobo-Dols | |
Sweden | Simon Ekman |
Bengt Bergman | |
Ronny Ohman | |
Anders Vikstrom | |
Taiwan | Chih-Hsin Yang |
Chao-Hua Chiu/Hsu-Ching Huang | |
Cheng-Ta Yang | |
Jian Su | |
Gee-Chen Chang/Tsung-Ying Yang | |
Te-Chun Hsia | |
Wu-Chou Su/Shang-Yin Wu | |
Chin-Chou Wang | |
Kang-Yun Lee | |
Sheng-Hao Lin | |
Chih-Bin Lin | |
Jih-Hsiang Lee | |
Chun-Yao Huang | |
UK | Samreen Ahmed |
Thomas Newsom-Davis | |
Shobhit Baijal | |
Juliet Brock/Kam Zaki | |
Jonathan Shamash | |
Dionysis Papadatos-Pastos | |
Pooja Jain | |
Melanie Jane Mackean | |
United States | Stephan DiSean Kendall |
Ian Anderson | |
Dan Costin | |
Richard Hall | |
Nicholas Campbell | |
Saad Khan/Jonathan Dowell | |
Sandeep Mashru | |
Smitha Menon | |
Ahmad Raza/Li Ge | |
Gregory J. Riely | |
Nagashree Seetharamu | |
Laura Stampleman | |
Janakiraman Subramanian | |
Donald B. Wender | |
Ronald B. Natale | |
Viola Zhu/Sai-Hong Ignatius Ou | |
Rachel Sanborn | |
Makenzi C. Evangelist |
NOTE. Where two names are listed, original investigator's name is followed by subsequent investigator's name.
James Chih-Hsin Yang
Honoraria: Boehringer Ingelheim, Roche, MSD, AstraZeneca, Novartis, Bristol Myers Squibb, Ono Pharmaceutical, Takeda, Lilly, Pfizer, Amgen (Inst), AstraZeneca/MedImmune (Inst), Boehringer Ingelheim (Inst), Dizal Pharma (Inst), Taiho Pharmaceutical (Inst), Pfizer (Inst), Takeda (Inst), Roche/Genentech (Inst), Daiichi Sankyo/Astra Zeneca (Inst), MSD Oncology (Inst), BeiGene (Inst), Gilead Sciences (Inst), Sanofi/Regeneron (Inst)
Consulting or Advisory Role: Boehringer Ingelheim, Novartis, AstraZeneca, Clovis Oncology, Lilly (Inst), MSD Oncology, Celgene, Bayer, Pfizer, Ono Pharmaceutical, Bristol Myers Squibb, Boehringer Ingelheim (Inst), Yuhan, Hansoh, Blueprint Medicines, Daiichi Sankyo, G1 Therapeutics, AbbVie, Takeda, Amgen, Incyte, GlaxoSmithKline (Inst), Amgen (Inst), Takeda (Inst), AstraZeneca (Inst), Novartis (Inst), MSD Oncology (Inst), Janssen Oncology (Inst), Merck KGaA (Inst), Daiichi Sankyo/Astra Zeneca (Inst), Puma Biotechnology (Inst), Gilead Sciences (Inst), Pfizer (Inst), Taiho Pharmaceutical (Inst), Bayer (Inst), Roche/Genentech (Inst), Sanofi (Inst)
Research Funding: AstraZeneca (Inst)
Travel, Accommodations, Expenses: Pfizer
Dae Ho Lee
Honoraria: AstraZeneca/MedImmune, Boehringer Ingelheim, Bristol Myers Squibb, Lilly, MSD, Novartis, Ono Pharmaceutical, Pfizer, Roche/Genentech, ST Cube, Takeda, AbbVie, Yuhan, ChongKeunDang Healthcare, Janssen
Consulting or Advisory Role: ST Cube, ABION
Filippo de Marinis
Consulting or Advisory Role: AstraZeneca, MSD Oncology, Bristol Myers Squibb, Roche/Genentech, Pfizer, Novartis, Takeda, Daichii Sankyo, Merck Serono
Eiji Iwama
Honoraria: AstraZeneca, Chugai Pharma, Novartis, Pfizer, Thermo Fisher Scientific, Daiichi Sankyo/Astra Zeneca
Takako Inoue
Speakers' Bureau: AstraZeneca, Chugai Pharma, Bristol Myers Squibb Japan, Ono Pharmaceutical, MSD
Jerónimo Rodríguez-Cid
Consulting or Advisory Role: Roche, Bristol Myers Squibb (Mexico), MSD Oncology, Takeda, Bayer, Pfizer, Janssen Oncology
Speakers' Bureau: MSD Oncology, Bristol Myers Squibb (Mexico), Roche, Boehringer Ingelheim, Novartis, Bayer, Lilly, AstraZeneca
Research Funding: MSD, Bristol Myers Squibb (Mexico), Roche, Celltrion, Lilly, BeiGene, AstraZeneca, Pfizer, Daiichi Sankyo/Astra Zeneca
Travel, Accommodations, Expenses: Roche, MSD Oncology, AstraZeneca, Boehringer Ingelheim
Cheng-Ta Yang
Consulting or Advisory Role: AstraZenica, Boehringer Ingelheim, Lilly, Merck, Ono, BMS
Speakers' Bureau: Novartis, AstraZenica, Boehringer Ingelheim, Lilly, MSD, Merck, Amgen, Johnson & Johnson, Roche, Ono, BMS, Chugai
Jianying Zhou
Consulting or Advisory Role: Hansoh Pharma
Research Funding: Hansoh Pharma (Inst)
Maurice Pérol
This author is a member of the Journal of Clinical Oncology Editorial Board. Journal policy recused the author from having any role in the peer review of this manuscript.
Consulting or Advisory Role: Lilly, Roche/Genentech, Pfizer, AstraZeneca, Merck Sharp & Dohme, Bristol Myers Squibb, Novartis, Amgen, Takeda, Sanofi, GlaxoSmithKline, Janssen Oncology, IPSEN, Eisai, Novocure, Daiichi Sankyo, Gilead Sciences
Research Funding: Takeda (Inst)
Travel, Accommodations, Expenses: AstraZeneca, Roche, Bristol Myers Squibb, Merck Sharp & Dohme, Pfizer, Takeda, Chugai Pharma
Ki Hyeong Lee
Consulting or Advisory Role: Bristol Myers Squibb, MSD, AstraZeneca, Pfizer, Lilly
Research Funding: Merck Serono, Merck Sharp & Dohme
David Vicente
Honoraria: AstraZeneca
Consulting or Advisory Role: Bristol Myers Squibb, MSD Oncology, Roche/Genentech, Pfizer, AstraZeneca, Boehringer Ingelheim, Gilead/Forty Seven, Novartis
Travel, Accommodations, Expenses: AstraZeneca
Eiki Ichihara
Honoraria: AstraZeneca, Takeda, Pfizer, Novartis, Chugai Pharma, MSD, Ono Pharmaceutical, Bristol Myers Squibb Japan, Merck
Research Funding: Janssen
Gregory J. Riely
Research Funding: Novartis (Inst), Roche/Genentech (Inst), Mirati Therapeutics (Inst), Merck (Inst), Takeda (Inst), Lilly (Inst), Pfizer (Inst)
Patents, Royalties, Other Intellectual Property: Patent application submitted covering pulsatile use of erlotinib to treat or prevent brain metastases (Inst)
Travel, Accommodations, Expenses: Bayer, Merck
Other Relationship: Pfizer, Roche/Genentech, Takeda, Mirati Therapeutics
Yiwen Luo
Employment: Merck Sharp & Dohme
Stock and Other Ownership Interests: Merck Sharp & Dohme
Travel, Accommodations, Expenses: Merck Sharp & Dohme
Diana Chirovsky
Employment: Merck
Stock and Other Ownership Interests: Merck
M. Catherine Pietanza
Employment: Merck
Stock and Other Ownership Interests: Merck Sharp & Dohme
Niyati Bhagwati
Employment: Merck
Stock and Other Ownership Interests: Merck
Travel, Accommodations, Expenses: Merck
Shun Lu
Leadership: Innovent Biologics, Inc
Consulting or Advisory Role: AstraZeneca, Pfizer, Boehringer Ingelheim, Hutchison MediPharma, Simcere, Zai Lab, GenomiCare, Yuhan, Roche, Menarini, InventisBio Co. Ltd
Speakers' Bureau: AstraZeneca, Roche, Hansoh Pharma, Hengrui Therapeutics
Research Funding: AstraZeneca (Inst), Hutchison MediPharma (Inst), BMS (Inst), Hengrui Therapeutics (Inst), BeiGene (Inst), Roche (Inst), Hansoh (Inst), Lilly Suzhou Pharmaceutical Co (Inst)
No other potential conflicts of interest were reported.
See accompanying Editorial, p. 4003
SUPPORT
Supported by Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc, Rahway, NJ, USA.
CLINICAL TRIAL INFORMATION
Contributor Information
Collaborators: Michael Joseph Boyer, Rina Hui, Mark Wong, Andrew Mant, Phillip Parente, Thomas John, Sagun Parakh, Gilberto Castro, Jr, Gustavo Werutsky, Sergio Jobim de Azevedo, Fabio Andre Franke, Joilda Batista De Almeida Rego, Pedro Rafael Martins De Marchi, Gustavo Dix Junqueira, Fernanda Maris Peria, Leandro Brust, Parneet Cheema, Mark Doherty, Ambika Parmar, Ines B. Menjak, Natasha B. Leighl, Jason Agulnik, Shun Lu, Zhigang Han, Jiuwei Cui, Li Zhang, Ying Cheng, Gongyan Chen, Helong Zhang, Yu Yao, Chengping Hu, Qiming Wang, Xin Zhang, Yong Zhang, Jianying Zhou, Kejing Ying, Yun Fan, Yan Wang, Ziping Wang, Jifeng Feng, YingYing Du, Lin Wu, Cheng Huang, Xiangdong Zhou, Maurice Perol, Julien Domont, Corinne Lamour, Julien Dutilh, Youssef Oulkhouir, Virginie Westeel, Delphine Carmier, Bruno Coudert, Aurelie Lagrange, Dominique Spaeth, Stanislas Ropert, Daniel C. Christoph, Jens Kern, Hans-Georg Kopp, Frank Griesinger, Rainer Wiewrodt, Martin Wermke, Claas Wesseler, Annette Mueller, Gunther Vogel, Victor Lee, Chung Man James Ho, Siu Hong Oscar Chan, Sing Hung Lo, Shi Feng Jonathan Nyaw, Yu Chung Jacky Li, Jair Bar, Maya Gottfried, Julia Dudnik, Alona Zer, Mor Moskovitz, Mirjana Wollner, Ofer Rotem, Sivan Shamai, Noam Asna, Mhameed Kamel, Silvia Novello, Francesco Di Costanzo, Laura Doni, Francesca Mazzoni, Francesco Ferrau, Filippo de Marinis, Giuseppe Tonini, Domenico Galetta, Francovito Piantedosi, Fabiana Vitiello, Keisuke Kirita, Kiyotaka Yoh, Toshiaki Takahashi, Yuichiro Ohe, Yoshihiro Hattori, Isamu Okamoto, Takayasu Kurata, Hiroshige Yoshioka, Hideo Saka, Masahide Oki, Terufumi Kato, Hiroshi Tanaka, Toru Kumagai, Takako Inoue, Toyoaki Hida, Yoshitsugu Horio, Shunsuke Teraoka, Eiki Ichihara, Kazuma Kishi, Hisashi Takaya, Daiya Takai, Toshiyuki Kozuki, Kazuo Kasahara, Yuichi Tambo, Yukio Hosomi, Masashi Kondo, Masao Ichiki, Hiroaki Takeoka, Emmanuel de la Mora Jimenez, Carlos Alberto Hernandez, Jeronimo Rafael Rodriguez Cid, Oscar Gerardo Arrieta Rodriguez, Ji-Youn Han, Young Joo Min, Dong-Wan Kim, Keunchil Park, Se-Hoon Lee, Ki Hyeong Lee, Jong-Seok Lee, Jin Hyoung Kang, Dae Ho Lee, Eun Kyung Cho, Enric Carcereny, Pilar Garrido Lopez, Margarita Majem Tarruella, Luis Paz-Ares Rodriguez, David Vicente Baz, Enriqueta Felip Font, Manuel Cobo-Dols, Simon Ekman, Bengt Bergman, Ronny Ohman, Anders Vikstrom, Chih-Hsin Yang, Chao-Hua Chiu, Hsu-Ching Huang, Cheng-Ta Yang, Jian Su, Gee-Chen Chang, Tsung-Ying Yang, Te-Chun Hsia, Wu-Chou Su, Shang-Yin Wu, Chin-Chou Wang, Kang-Yun Lee, Sheng-Hao Lin, Chih-Bin Lin, Jih-Hsiang Lee, Chun-Yao Huang, Samreen Ahmed, Thomas Newsom-Davis, Shobhit Baijal, Juliet Brock, Kam Zaki, Jonathan Shamash, Dionysis Papadatos-Pastos, Pooja Jain, Melanie Jane Mackean, Stephan DiSean Kendall, Ian Anderson, Dan Costin, Richard Hall, Nicholas Campbell, Saad Khan, Jonathan Dowell, Sandeep Mashru, Smitha Menon, Ahmad Raza, Li Ge, Gregory J. Riely, Nagashree Seetharamu, Laura Stampleman, Janakiraman Subramanian, Donald B. Wender, Ronald B. Natale, Viola Zhu, Sai-Hong Ignatius Ou, Rachel Sanborn, and Makenzi C. Evangelist
DATA SHARING STATEMENT
Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc, Rahway, NJ, USA (MSD), is committed to providing qualified scientific researchers access to anonymized data and clinical study reports from the company's clinical trials for the purpose of conducting legitimate scientific research. MSD is also obligated to protect the rights and privacy of trial participants and, as such, has a procedure in place for evaluating and fulfilling requests for sharing company clinical trial data with qualified external scientific researchers. The MSD data sharing website (available at http://engagezone.msd.com/ds_documentation.php) outlines the process and requirements for submitting a data request. Applications will be promptly assessed for completeness and policy compliance. Feasible requests will be reviewed by a committee of MSD subject matter experts to assess the scientific validity of the request and the qualifications of the requestors. In line with data privacy legislation, submitters of approved requests must enter into a standard data-sharing agreement with MSD before data access is granted. Data will be made available for request after product approval in the United States and EU or after product development is discontinued. There are circumstances that may prevent MSD from sharing requested data, including country- or region-specific regulations. If the request is declined, it will be communicated to the investigator. Access to genetic or exploratory biomarker data requires a detailed, hypothesis-driven statistical analysis plan that is collaboratively developed by the requestor and MSD subject matter experts; after approval of the statistical analysis plan and execution of a data-sharing agreement, MSD will either perform the proposed analyses and share the results with the requestor or construct biomarker covariates and add them to a file with clinical data that is uploaded to an analysis portal so that the requestor can perform the proposed analyses.
AUTHOR CONTRIBUTIONS
Conception and design: James Chih-Hsin Yang, Li Zhang, Ki Hyeong Lee, Gregory J. Riely, Yiwen Luo, M. Catherine Pietanza
Administrative support: Shun Lu
Provision of study materials or patients: Dae Ho Lee, Jong-Seok Lee, Yun Fan, Filippo de Marinis, Eiji Iwama, Jerónimo Rodríguez-Cid, Cheng-Ta Yang, Emmanuel de la Mora Jimenez, Jianying Zhou, Maurice Pérol, Ki Hyeong Lee, Eiki Ichihara, Niyati Bhagwati, Shun Lu
Collection and assembly of data: James Chih-Hsin Yang, Dae Ho Lee, Jong-Seok Lee, Yun Fan, Eiji Iwama, Takako Inoue, Jerónimo Rodríguez-Cid, Li Zhang, Cheng-Ta Yang, Emmanuel de la Mora Jimenez, Jianying Zhou, Maurice Pérol, Ki Hyeong Lee, Eiki Ichihara, Yiwen Luo, Niyati Bhagwati, Shun Lu
Data analysis and interpretation: James Chih-Hsin Yang, Dae Ho Lee, Filippo de Marinis, Maurice Pérol, Ki Hyeong Lee, David Vicente, Gregory J. Riely, Yiwen Luo, Diana Chirovsky, M. Catherine Pietanza, Niyati Bhagwati, Shun Lu
Manuscript writing: All authors
Final approval of manuscript: All authors
Accountable for all aspects of the work: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Phase III KEYNOTE-789 Study of Pemetrexed and Platinum With or Without Pembrolizumab for Tyrosine Kinase Inhibitor‒Resistant, EGFR–Mutant, Metastatic Nonsquamous Non–Small Cell Lung Cancer
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/authors/author-center.
Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).
James Chih-Hsin Yang
Honoraria: Boehringer Ingelheim, Roche, MSD, AstraZeneca, Novartis, Bristol Myers Squibb, Ono Pharmaceutical, Takeda, Lilly, Pfizer, Amgen (Inst), AstraZeneca/MedImmune (Inst), Boehringer Ingelheim (Inst), Dizal Pharma (Inst), Taiho Pharmaceutical (Inst), Pfizer (Inst), Takeda (Inst), Roche/Genentech (Inst), Daiichi Sankyo/Astra Zeneca (Inst), MSD Oncology (Inst), BeiGene (Inst), Gilead Sciences (Inst), Sanofi/Regeneron (Inst)
Consulting or Advisory Role: Boehringer Ingelheim, Novartis, AstraZeneca, Clovis Oncology, Lilly (Inst), MSD Oncology, Celgene, Bayer, Pfizer, Ono Pharmaceutical, Bristol Myers Squibb, Boehringer Ingelheim (Inst), Yuhan, Hansoh, Blueprint Medicines, Daiichi Sankyo, G1 Therapeutics, AbbVie, Takeda, Amgen, Incyte, GlaxoSmithKline (Inst), Amgen (Inst), Takeda (Inst), AstraZeneca (Inst), Novartis (Inst), MSD Oncology (Inst), Janssen Oncology (Inst), Merck KGaA (Inst), Daiichi Sankyo/Astra Zeneca (Inst), Puma Biotechnology (Inst), Gilead Sciences (Inst), Pfizer (Inst), Taiho Pharmaceutical (Inst), Bayer (Inst), Roche/Genentech (Inst), Sanofi (Inst)
Research Funding: AstraZeneca (Inst)
Travel, Accommodations, Expenses: Pfizer
Dae Ho Lee
Honoraria: AstraZeneca/MedImmune, Boehringer Ingelheim, Bristol Myers Squibb, Lilly, MSD, Novartis, Ono Pharmaceutical, Pfizer, Roche/Genentech, ST Cube, Takeda, AbbVie, Yuhan, ChongKeunDang Healthcare, Janssen
Consulting or Advisory Role: ST Cube, ABION
Filippo de Marinis
Consulting or Advisory Role: AstraZeneca, MSD Oncology, Bristol Myers Squibb, Roche/Genentech, Pfizer, Novartis, Takeda, Daichii Sankyo, Merck Serono
Eiji Iwama
Honoraria: AstraZeneca, Chugai Pharma, Novartis, Pfizer, Thermo Fisher Scientific, Daiichi Sankyo/Astra Zeneca
Takako Inoue
Speakers' Bureau: AstraZeneca, Chugai Pharma, Bristol Myers Squibb Japan, Ono Pharmaceutical, MSD
Jerónimo Rodríguez-Cid
Consulting or Advisory Role: Roche, Bristol Myers Squibb (Mexico), MSD Oncology, Takeda, Bayer, Pfizer, Janssen Oncology
Speakers' Bureau: MSD Oncology, Bristol Myers Squibb (Mexico), Roche, Boehringer Ingelheim, Novartis, Bayer, Lilly, AstraZeneca
Research Funding: MSD, Bristol Myers Squibb (Mexico), Roche, Celltrion, Lilly, BeiGene, AstraZeneca, Pfizer, Daiichi Sankyo/Astra Zeneca
Travel, Accommodations, Expenses: Roche, MSD Oncology, AstraZeneca, Boehringer Ingelheim
Cheng-Ta Yang
Consulting or Advisory Role: AstraZenica, Boehringer Ingelheim, Lilly, Merck, Ono, BMS
Speakers' Bureau: Novartis, AstraZenica, Boehringer Ingelheim, Lilly, MSD, Merck, Amgen, Johnson & Johnson, Roche, Ono, BMS, Chugai
Jianying Zhou
Consulting or Advisory Role: Hansoh Pharma
Research Funding: Hansoh Pharma (Inst)
Maurice Pérol
This author is a member of the Journal of Clinical Oncology Editorial Board. Journal policy recused the author from having any role in the peer review of this manuscript.
Consulting or Advisory Role: Lilly, Roche/Genentech, Pfizer, AstraZeneca, Merck Sharp & Dohme, Bristol Myers Squibb, Novartis, Amgen, Takeda, Sanofi, GlaxoSmithKline, Janssen Oncology, IPSEN, Eisai, Novocure, Daiichi Sankyo, Gilead Sciences
Research Funding: Takeda (Inst)
Travel, Accommodations, Expenses: AstraZeneca, Roche, Bristol Myers Squibb, Merck Sharp & Dohme, Pfizer, Takeda, Chugai Pharma
Ki Hyeong Lee
Consulting or Advisory Role: Bristol Myers Squibb, MSD, AstraZeneca, Pfizer, Lilly
Research Funding: Merck Serono, Merck Sharp & Dohme
David Vicente
Honoraria: AstraZeneca
Consulting or Advisory Role: Bristol Myers Squibb, MSD Oncology, Roche/Genentech, Pfizer, AstraZeneca, Boehringer Ingelheim, Gilead/Forty Seven, Novartis
Travel, Accommodations, Expenses: AstraZeneca
Eiki Ichihara
Honoraria: AstraZeneca, Takeda, Pfizer, Novartis, Chugai Pharma, MSD, Ono Pharmaceutical, Bristol Myers Squibb Japan, Merck
Research Funding: Janssen
Gregory J. Riely
Research Funding: Novartis (Inst), Roche/Genentech (Inst), Mirati Therapeutics (Inst), Merck (Inst), Takeda (Inst), Lilly (Inst), Pfizer (Inst)
Patents, Royalties, Other Intellectual Property: Patent application submitted covering pulsatile use of erlotinib to treat or prevent brain metastases (Inst)
Travel, Accommodations, Expenses: Bayer, Merck
Other Relationship: Pfizer, Roche/Genentech, Takeda, Mirati Therapeutics
Yiwen Luo
Employment: Merck Sharp & Dohme
Stock and Other Ownership Interests: Merck Sharp & Dohme
Travel, Accommodations, Expenses: Merck Sharp & Dohme
Diana Chirovsky
Employment: Merck
Stock and Other Ownership Interests: Merck
M. Catherine Pietanza
Employment: Merck
Stock and Other Ownership Interests: Merck Sharp & Dohme
Niyati Bhagwati
Employment: Merck
Stock and Other Ownership Interests: Merck
Travel, Accommodations, Expenses: Merck
Shun Lu
Leadership: Innovent Biologics, Inc
Consulting or Advisory Role: AstraZeneca, Pfizer, Boehringer Ingelheim, Hutchison MediPharma, Simcere, Zai Lab, GenomiCare, Yuhan, Roche, Menarini, InventisBio Co. Ltd
Speakers' Bureau: AstraZeneca, Roche, Hansoh Pharma, Hengrui Therapeutics
Research Funding: AstraZeneca (Inst), Hutchison MediPharma (Inst), BMS (Inst), Hengrui Therapeutics (Inst), BeiGene (Inst), Roche (Inst), Hansoh (Inst), Lilly Suzhou Pharmaceutical Co (Inst)
No other potential conflicts of interest were reported.
REFERENCES
- 1.Shields MD, Marin-Acevedo JA, Pellini B: Immunotherapy for advanced non-small cell lung cancer: A decade of progress. Am Soc Clin Oncol Educ Book 41:1-23, 2021 [DOI] [PubMed] [Google Scholar]
- 2.Hendriks LE, Kerr KM, Menis J, et al. : Oncogene-addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 34:339-357, 2023 [DOI] [PubMed] [Google Scholar]
- 3.Lee CK, Man J, Lord S, et al. : Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer—A meta-analysis. J Thorac Oncol 12:403-407, 2017 [DOI] [PubMed] [Google Scholar]
- 4.Lee CK, Man J, Lord S, et al. : Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non–small cell lung carcinoma: A systematic review and meta-analysis. JAMA Oncol 4:210-216, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Shi C, Wang Y, Xue J, et al. : Immunotherapy for EGFR-mutant advanced non-small-cell lung cancer: Current status, possible mechanisms and application prospects. Front Immunol 13:940288, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Gainor JF, Shaw AT, Sequist LV, et al. : EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: A retrospective analysis. Clin Cancer Res 22:4585-4593, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.National Comprehensive Cancer Network (NCCN) : NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines): Non-small Cell Lung Cancer. Version 3.2023, 2023 [Google Scholar]
- 8.Wu L, Ke L, Zhang Z, et al. : Development of EGFR TKIs and options to manage resistance of third-generation EGFR TKI osimertinib: Conventional ways and immune checkpoint inhibitors. Front Oncol 10:602762, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Qian X, Guo X, Li T, et al. : Efficacy of immune checkpoint inhibitors in EGFR-mutant NSCLC patients with EGFR-TKI resistance: A systematic review and meta-analysis. Front Pharmacol 13:926890, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Reck M, Mok TSK, Nishio M, et al. : Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): Key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir Med 7:387-401, 2019 [DOI] [PubMed] [Google Scholar]
- 11.To KKW, Fong W, Cho WCS: Immunotherapy in treating EGFR-mutant lung cancer: Current challenges and new strategies. Front Oncol 11:635007, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Reck M, Rodriguez-Abreu D, Robinson AG, et al. : Pembrolizumab versus chemotherapy for PD-L1-positive non–small-cell lung cancer. N Engl J Med 375:1823-1833, 2016 [DOI] [PubMed] [Google Scholar]
- 13.Mok TSK, Wu YL, Kudaba I, et al. : Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomised, open-label, controlled, phase 3 trial. Lancet 393:1819-1830, 2019 [DOI] [PubMed] [Google Scholar]
- 14.Gandhi L, Rodriguez-Abreu D, Gadgeel S, et al. : Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med 378:2078-2092, 2018 [DOI] [PubMed] [Google Scholar]
- 15.Paz-Ares L, Luft A, Vicente D, et al. : Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med 379:2040-2051, 2018 [DOI] [PubMed] [Google Scholar]
- 16.Seymour L, Bogaerts J, Perrone A, et al. : iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncology 18:e143-e152, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Nishio M, Barlesi F, West H, et al. : Atezolizumab plus chemotherapy for first-line treatment of nonsquamous NSCLC: Results from the randomized phase 3 IMpower132 trial. J Thorac Oncol 16:653-664, 2021 [DOI] [PubMed] [Google Scholar]
- 18.Attili I, Passaro A, Corvaja C, et al. : Immune checkpoint inhibitors in EGFR-mutant non-small cell lung cancer: A systematic review. Cancer Treat Rev 119:102602, 2023 [DOI] [PubMed] [Google Scholar]
- 19.Dong ZY, Zhang JT, Liu SY, et al. : EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. Oncoimmunology 6:e1356145, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wan C, Zhang Y, Liu P, et al. : Efficacy and safety of anti-programmed cell death protein 1 antibody combination therapy in patients with advanced experienced epidermal growth factor receptor-tyrosine kinase inhibitor-resistant lung adenocarcinoma: A retrospective cohort study. J Thorac Dis 15:5648-5657, 2023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Park S, Kim TM, Han JY, et al. : A phase 3, randomized study of atezolizumab plus bevacizumab and chemotherapy in patients with EGFR or ALK mutated in non-small cell lung cancer (ATTLAS, KCSG-LU19-04). J Clin Oncol 42:1241-1251, 2024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Arrieta O, Barron F, Ramirez-Tirado LA, et al. : Efficacy and safety of pembrolizumab plus docetaxel vs docetaxel alone in patients with previously treated advanced non-small cell lung cancer: The PROLUNG phase 2 randomized clinical trial. JAMA Oncol 6:856-864, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Gadgeel S, Dziubek K, Nagasaka M, et al. : OA09.03 Pembrolizumab in combination with platinum-based chemotherapy in recurrent EGFR/ALK-positive non-small cell lung cancer (NSCLC). J Thorac Oncol 16:S863, 2021 [Google Scholar]
- 24.Mok T, Nakagawa K, Park K, et al. : Nivolumab plus chemotherapy in epidermal growth factor receptor–mutated metastatic non–small-cell lung cancer after disease progression on epidermal growth factor receptor tyrosine kinase inhibitors: Final results of CheckMate 722. J Clin Oncol 42:1252-1264, 2024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Lu S, Wu L, Jian H, et al. : Sintilimab plus chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer with disease progression after EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): Second interim analysis from a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Respir Med 11:624-636, 2023 [DOI] [PubMed] [Google Scholar]
- 26.Lu S, Wu L, Jian H, et al. : Sintilimab plus bevacizumab biosimilar IBI305 and chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer who progressed on EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): First interim results from a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol 23:1167-1179, 2022 [DOI] [PubMed] [Google Scholar]
- 27.Chen F, Chen N, Yu Y, et al. : Efficacy and safety of epidermal growth factor receptor (EGFR) inhibitors plus antiangiogenic agents as first-line treatments for patients with advanced EGFR-mutated non-small cell lung cancer: A meta-analysis. Front Oncol 10:904, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Remon J, Lacas B, Herbst R, et al. : ANtiangiogenic second-line lung cancer meta-analysis on individual patient data in non-small cell lung cancer: ANSELMA. Eur J Cancer 166:112-125, 2022 [DOI] [PubMed] [Google Scholar]
- 29.Nakagawa K, Garon EB, Seto T, et al. : Ramucirumab plus erlotinib in patients with untreated, EGFR-mutated, advanced non-small-cell lung cancer (RELAY): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 20:1655-1669, 2019 [DOI] [PubMed] [Google Scholar]
- 30.Fu K, Xie F, Wang F, et al. : Therapeutic strategies for EGFR-mutated non-small cell lung cancer patients with osimertinib resistance. J Hematol Oncol 15:173, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Han R, Guo H, Shi J, et al. : Tumour microenvironment changes after osimertinib treatment resistance in non-small cell lung cancer. Eur J Cancer 189:112919, 2023 [DOI] [PubMed] [Google Scholar]
- 32.Jiang XM, Xu YL, Huang MY, et al. : Osimertinib (AZD9291) decreases programmed death ligand-1 in EGFR-mutated non-small cell lung cancer cells. Acta Pharmacol Sin 38:1512-1520, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc, Rahway, NJ, USA (MSD), is committed to providing qualified scientific researchers access to anonymized data and clinical study reports from the company's clinical trials for the purpose of conducting legitimate scientific research. MSD is also obligated to protect the rights and privacy of trial participants and, as such, has a procedure in place for evaluating and fulfilling requests for sharing company clinical trial data with qualified external scientific researchers. The MSD data sharing website (available at http://engagezone.msd.com/ds_documentation.php) outlines the process and requirements for submitting a data request. Applications will be promptly assessed for completeness and policy compliance. Feasible requests will be reviewed by a committee of MSD subject matter experts to assess the scientific validity of the request and the qualifications of the requestors. In line with data privacy legislation, submitters of approved requests must enter into a standard data-sharing agreement with MSD before data access is granted. Data will be made available for request after product approval in the United States and EU or after product development is discontinued. There are circumstances that may prevent MSD from sharing requested data, including country- or region-specific regulations. If the request is declined, it will be communicated to the investigator. Access to genetic or exploratory biomarker data requires a detailed, hypothesis-driven statistical analysis plan that is collaboratively developed by the requestor and MSD subject matter experts; after approval of the statistical analysis plan and execution of a data-sharing agreement, MSD will either perform the proposed analyses and share the results with the requestor or construct biomarker covariates and add them to a file with clinical data that is uploaded to an analysis portal so that the requestor can perform the proposed analyses.