We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Short CommunicationOpen Accesscc iconby iconnc iconnd icon

Real-world relationship of early end points to survival end points in patients with resectable non-small-cell lung cancer

    Eric Nadler

    Charles Sammons Cancer Center, Baylor University Medical Center, US Oncology Network, Dallas, TX 75246, USA

    , , , ,
    John R Penrod

    Bristol Myers Squibb, Lawrenceville, NJ 08648, USA

    ,
    Joseph Fiore

    Bristol Myers Squibb, Lawrenceville, NJ 08648, USA

    &
    Lien Vo

    *Author for correspondence:

    E-mail Address: lien.vo@bms.com

    Bristol Myers Squibb, Lawrenceville, NJ 08648, USA

    Published Online:https://doi.org/10.2217/fon-2023-0170

    Abstract

    Aim: Pathologic response has been shown to be a promising surrogate for survival in non-small-cell lung cancer. We examined the real-world relationship between these end  points in patients with resectable stage IB-IIIA non-small-cell lung cancer receiving neoadjuvant chemotherapy/chemoradiotherapy (CT/CRT). Methods: Electronic health records/medical charts were analyzed. Overall and event-free survival (OS/EFS) were assessed by Kaplan-Meier stratified by pathologic response. Associations between the end  points were assessed by Cox analyses. Results: A total of 425 patients were selected for the study; 147 and 278 received CT and CRT, respectively. Pathologic complete response (pCR) was associated with longer OS (adjusted HR = 0.50; 95% CI: 0.29–0.85) and EFS (adjusted HR = 0.44; 95% CI: 0.28–0.68) versus no pCR, and EFS was associated with OS (HR = 0.51, 95% CI: 0.38, 0.69). Conclusion: In patients receiving neoadjuvant CT/CRT, pCR and EFS were associated with improved survival in this real-world dataset.

    Plain language summary – A faster way to see if new cancer drugs work

    Many patients with early-stage lung cancer have their cancer come back in 2–5 years after treatment, and when it returns it is often incurable. This has been true for many cancer drugs over many years. However, developing new and better cancer drugs is difficult. In drug trials, it takes years to see if the patients live longer. Disappearance of cancer tissue can show earlier if a cancer drug is working. This is called ‘pathological response’.

    We studied a large number of patients with early lung cancer. These patients were treated at community cancer practices. Some of these patients had pathological response. Those patients lived longer and/or it took longer for the cancer to come back. The study showed that pathological response is a faster way to see if new cancer drugs work.

    Tweetable abstract

    In patients with resectable stage IB-IIIA non-small-cell lung cancer receiving neoadjuvant chemotherapy/chemoradiotherapy at community oncology practices, complete pathologic response and event-free survival were both associated with improved survival.

    Lung cancer is the leading cause of cancer death in USA, with 80–85% of cases being non-small-cell lung cancer (NSCLC). At initial diagnosis, 26% of patients present with stage I disease, 8.3% with stage II, 27.6% with stage III, and 38.1% with stage IV disease [1]. For many patients with non-metastatic NSCLC, surgery with curative intent has been the gold standard for many years. Despite surgery, 30 to 55% of patients with NSCLC eventually develop recurrence within 2–5 years and die of their disease [2–6]. Patients with stage IB-IIIA NSCLC still represent a population with a high unmet need, with a 5-year survival rate ranging from 36 to 66% [7].

    Historically, neoadjuvant or adjuvant chemotherapy with or without radiotherapy have been recommended for patients at higher risk of recurrence [8,9]. These treatment strategies have been associated with slower recurrence and modestly prolonged survival among patients with stage IB-IIIA disease [10–13]. However, both strategies have only shown approximately 5% overall survival (OS) absolute risk reduction at 5 years compared with surgery alone [7].

    Typically, the investigation of improvement in outcomes for patients with resectable lung cancers has been challenging, limiting the development and approval of new drugs. Phase III trials with conventional primary end points (e.g., OS) can take up to 10 years to complete the accrual and required follow-up [7,14]. Over the years, these trials have struggled to accrue, leaving several underpowered for their event-free survival (EFS) and OS end points [7]. In addition, these large and lengthy trials may be confounded by the use of additional post-operative interventions based on local clinical practice, including chemotherapy or radiotherapy to address residual disease. Hence, there is a need for early or intermediate end points that both isolate the effect of novel neoadjuvant interventions and expedite development and approval of promising treatments for patients with resectable NSCLC.

    Over the last decade, pCR and MPR have been proposed as early end points for prediction of long-term clinical benefit, such as EFS and OS among patients with NSCLC, and are increasingly being included in neoadjuvant clinical trials [15–18]. Prior research has found that pathologic response is predictive of OS and EFS [19,20]. However, heterogeneous definitions of pCR, low pCR rates and infrequent reporting of MPR in clinical trials present a major challenge while evaluating the outcomes in patients with NSCLC [15,17,21,22].

    The purpose of this study is to generate real-world data characterizing the relationship between early end points and survival-based outcomes and to describe the patient profiles and neoadjuvant treatment patterns in patients with resectable NSCLC (stage IB [tumor ≥4 cm]-IIIA) [23,24] treated in the US community oncology setting.

    Materials & methods

    Study design & population

    This is a retrospective cohort study including patients with a diagnosis of stage IB (tumor ≥4 cm), II, or IIIA NSCLC receiving neoadjuvant treatment at practices within The US Oncology Network that utilize the iKnowMed™ electronic health records (iKM EHR). The US Oncology Network consists of approximately 1400 physicians in more than 500 sites of care across 40 states in the USA, representing approximately 12% of US patients newly diagnosed with cancer [25]. Data were obtained via programmatic database abstraction from the EHR and supplemented with chart review. Vital status was confirmed with data from the Limited Access Death Master File (LADMF) and chart review. Eligible patients were ≥18 years of age with a diagnosis of stage IB (tumor ≥4 cm)-IIIA NSCLC1 or stage II–IIIB2 who initiated neoadjuvant treatment between 1 January 2008 and 31 January 2019 (defined as the patient identification period) with an intention of surgery and with ≥2 visits or a record of death after the index date (date of neoadjuvant treatment initiation) through 31 July 2019, which was the end of the study observation period. Patients were followed through the last visit, death or end of the study period, whichever came first. Patients were excluded if they were enrolled in clinical trials for non-metastatic NSCLC at any time during the study period or if they were treated for a documented primary cancer other than NSCLC during the study period. Patients with documented EGFR mutations or ALK translocation at diagnosis were excluded. Patients with EGFR mutations/ALK translocations are different from patients without. These differences include patient demographics, clinical characteristics and outcomes. In particular, given the adoption and efficacy of EGFR and ALK therapies in the metastatic/recurrent setting, outcomes for these patients would differ from wild-type patients, which could confound the analysis.

    Statement of ethics

    Institutional Review Board approval was obtained for the study. All data were handled in compliance with the Health Insurance Portability and Accountability Act of 1996 and the Health Information Technology for Economic and Clinical Health Act.

    Study outcomes

    The study explored the following outcomes: pCR, MPR, EFS and OS. These outcomes were analyzed for the overall study population and by type of neoadjuvant therapy (chemotherapy only, chemoradiotherapy). pCR was defined as no viable tumor cells in resected tumor tissue and lymph node tissue as reported in the pathology report or physician notes from the chart review of the index surgery, pathologic stage of pT0 pN0 or ypT0 ypN0. MPR was defined as ≤10% residual tumor tissue in the resected lung specimen. Patients with no surgery and with no information on pCR were classified as non-pCR. pCR was calculated for all patients who received neoadjuvant treatment with an intention of surgery. The as-treated approach is important, as isolating only patients that undergo surgery would introduce bias and overinflate pathologic response rates, excluding several real-world situations where patients may initiate treatment but discontinue (e.g. toxicity) or not be able to make it to surgery (e.g. tumor may progress while awaiting resection) and have worse outcomes than going straight to surgery.

    Progression or recurrent disease was defined by the availability of provider-recorded evidence of recurrence or progression in a scan report or progress note.

    EFS was estimated as the interval from the index date until the earliest date of progression of disease precluding surgery, progression or recurrence after surgery or death due to any cause. Patients who did not undergo surgery for any reason other than progression were considered to have had an event at progression or death. Patients without relapse, recurrence, progression or death were censored on the study end date or the last visit date, whichever occurred first, available in the database.

    OS was defined as the time interval between the index date and the date of death as documented in the LADMF or the iKM EHR database. Patients who did not die were censored on the study end date or the last visit date available in the database, whichever occurred first.

    Statistical analysis

    Descriptive analyses were conducted to summarize the baseline patient characteristics (clinical and treatment) for the study cohort population. The baseline period was defined as a 6-month period prior to the index date. These characteristics were presented for the overall study population and by type of neoadjuvant therapy (chemotherapy only, chemoradiotherapy). Continuous variables were described by means, standard deviations, medians and ranges, and categorical study variables were reported as frequencies (counts and percentages). Kaplan-Meier survival curves were generated for the outcome of OS/EFS for patients with and without pCR/MPR. An unadjusted Cox analysis was used to calculate HRs with 95% CIs for the association between pCR/MPR and OS/EFS in the overall population. A Cox proportional hazards model controlling for demographic and clinical characteristics (e.g., age, gender, disease stage, histology, site) was then conducted to describe the association between pCR and OS. The variables with a p-value less than or equal to 0.20 in Cox univariate analyses were included in the multivariable analyses. Landmark analyses were performed according to an event status at 3, 6, 12 and 24 months to identify survival outcomes, and unadjusted HR for OS was calculated. Additionally, EFS was used as a time-dependent covariate in a Cox regression on OS. As a patient-level analysis of the association between EFS and OS, event time versus survival time was plotted for each participant and the overall patient-level correlation between EFS and OS was calculated (Kendall's Tau rank correlation).

    Results

    Study population

    A total of 858 eligible patients with NSCLC were identified in the structured dataset. After further inclusion/exclusion criteria were applied during the chart review, 425 patients were eligible for this analysis (overall population); among these patients, 278 and 147 were treated with chemoradiotherapy and chemotherapy, respectively (Figure 1).

    Figure 1. Study attrition and sample size.

    AJCC: American Joint Committee on cancer; ALK: Anaplastic lymphoma kinase; EGFR: Epidermal growth factor receptor; NSCLC: Non-small-cell lung cancer; TNM: Tumor node metastasis.

    Demographics & baseline characteristics

    The median ages of patients treated with neoadjuvant chemotherapy and chemoradiotherapy were 67 years (range: 41, 88) and 66 years (range: 27, 89), respectively (Table 1). Most patients among both treatment cohorts had stage IIIA disease at index. Among the patients treated with chemotherapy, most patients had stage III disease (67.3%), followed by stage IIA (16.3%), stage IIB (15.0%) and stage IB (1.4%). Among the patients treated with chemoradiotherapy, most patients had stage III disease (65.5%), followed by stage IIB (21.9%), IIA (9.0%) and IB (3.6%). The proportion of patients with an ECOG PS score of 0 or 1 were numerically similar between patients treated with chemotherapy and chemoradiotherapy. The median (range) follow-up was 18.8 months (0.4, 135.6) among these patients. The most common comorbidities overall were hypertension (12.0%), ischemic disease (6.4%), pulmonary disease/asthma (6.4%) and diabetes (5.4%). More than 42% of the patients had at least 24 months of follow-up.

    Table 1. Baseline demographics and clinical characteristics of patients with non-small-cell lung cancer initiated on neoadjuvant therapy.
    VariableNeoadjuvant therapy
     Overall (N = 425)Chemotherapy (N = 147)Chemoradiotherapy (N = 278)
    Age at index (years)   
      Mean (SD)65.4 (10)66.3 (10.1)65 (9.9)
      Median (Min, Max)66 (27, 89)67 (41, 88)66 (27, 89)
    Age group - n (%)   
      <45 years12 (2.8)4 (2.7)8 (2.9)
      45 – 54 years45 (10.6)15 (10.2)30 (10.8)
      55 – 64 years128 (30.1)41 (27.9)87 (31.3)
      65 – 74 years159 (37.4)55 (37.4)104 (37.4)
      ≥75 years81 (19.1)32 (21.8)49 (17.6)
    Gender - n (%)   
      Female193 (45.4)62 (42.2)131 (47.1)
      Male232 (54.6)85 (57.8)147 (52.9)
    Race - n (%)   
      Black or African–American22 (5.2)9 (6.1)13 (4.7)
      Caucasian286 (67.3)94 (63.9)192 (69.1)
      Suppressed/other31 (7.3)13 (8.8)18 (6.5)
      Not documented86 (20.2)31 (21.1)55 (19.8)
    Practice region - n (%)   
      Midwest43 (10.1)5 (3.4)38 (13.7)
      Northeast80 (18.8)24 (16.3)56 (20.1)
      South167 (39.3)72 (49)95 (34.2)
      West135 (31.8)46 (31.3)89 (32)
    Body mass index (kg/m2) at index - n (%)  
      Underweight (<18.5)13 (3.1)1 (0.7)12 (4.3)
      Normal (≥18.5 and <25.0)138 (32.5)49 (33.3)89 (32)
      Overweight (≥25 and <30.0)151 (35.5)54 (36.7)97 (34.9)
      Obese (≥30.0)123 (28.9)43 (29.3)80 (28.8)
    Smoking status at index - n (%)  
      Never smoked3 (0.7)2 (1.4)1 (0.4)
      Current smoker26 (6.1)4 (2.7)22 (7.9)
      Former smoker263 (61.9)94 (63.9)169 (60.8)
      No information133 (31.3)47 (32)86 (30.9)
    Additional visits with practice - n (%)  
      Mean (SD)30.9 (22.3)30.7 (26.8)31 (19.6)
      Median (Min, Max)25 (1, 171)22 (1, 171)27 (2, 120)

    †Suppressed: Information was not reported for variables with n≤5 for HIPAA reasons. Other included Asian race, declined to specify, other and unknown.

    HIPAA: Health Insurance Portability and Accountability Act of 1996; SD: Standard deviation.

    Treatment characteristics

    Among the patients treated with neoadjuvant chemotherapy, the median (range) duration of treatment was 9.1 (0.1, 28.3) weeks or 3 (1, 15) cycles (Table 2). All except for one patient (gemcitabine) received platinum-based therapies; 53.7% received carboplatin-based and 45.6% received cisplatin-based. The most common neoadjuvant regimens were paclitaxel + carboplatin (34.7%), cisplatin + pemetrexed (17.7%) and carboplatin + pemetrexed (8.8%).

    Table 2. Treatment characteristics of patients with non-small-cell lung cancer initiated on neoadjuvant therapy.
     Neoadjuvant therapy
     Overall (N = 425)Chemotherapy (N = 147)Chemoradiotherapy (N = 278)
    Radiation therapy   
      Concurrent  264 (95)
      Sequential  1 (0.4)
      Not documented  13 (4.7)
    Radiation therapy - Type   
      EBRT  62 (22.3)
      IMRT  51 (18.3)
      SBRT/SABR  2 (0.7)
      Other  163 (58.6)
    Radiation therapy - dose Gy   
      Patients with available data  225
      Mean (SD)  52 (10.87)
      Median (range)  8.4 (0.67, 71.20)
    Neoadjuvant treatment regimen – n (%)   
      Carboplatin-based243 (57.2)79 (53.7)164 (59.0)
      Cisplatin-based179 (42.1)67 (45.6)112 (40.3)
      Paclitaxel + carboplatin209 (49.2)51 (34.7)158 (56.8)
      Pemetrexed + cisplatin32 (7.5)26 (17.7)6 (2.2)
      Docetaxel + cisplatin15 (3.5)14 (9.5)1 (0.4)
      Gemcitabine + cisplatin15 (3.5)14 (9.5)1 (0.4)
      Vinorelbine + cisplatin9 (2.1)8 (5.4)1 (0.4)
      Others145 (34.1)34 (23.1)111 (39.9)
    Others broken out: - n (%)   
    Total others14534111
      Cisplatin + Etoposide105 (72.4)5 (14.7)100 (90.1)
      Carboplatin + Pemetrexed17 (11.7)13 (38.2)4 (3.6)
      Carboplatin + Other7 (4.8)7 (20.6)0 (0)
      Carboplatin + Gemcitabine5 (3.4)5 (14.7)0 (0)
      Carboplatin + Etoposide3 (2.1)2 (5.9)1 (0.9)
      Carboplatin2 (1.4)1 (2.9)1 (0.9)
      Cisplatin + Other1 (0.7)0 (0)1 (0.9)
      Cisplatin + Other + Pemetrexed1 (0.7)0 (0)1 (0.9)
      Cisplatin + Paclitaxel1 (0.7)0 (0)1 (0.9)
      Docetaxel1 (0.7)0 (0)1 (0.9)
      Gemcitabine1 (0.7)1 (2.9)0 (0)
      Paclitaxel1 (0.7)0 (0)1 (0.9)
    Duration of neoadjuvant therapy (weeks)   
      Mean (SD)8.2 (4.8)8.6 (4.8)7.9 (4.8)
      Median (min, max)6.4 (0.1, 32.9)9.1 (0.1, 28.3)6.3 (1.0, 32.9)
    Number of cycles   
      Mean (SD)3.84 (2.28)3.46 (1.66)4.04 (2.53)
      Median (range)3 (1, 16)3 (1, 15)3 (1, 16)

    †Denominators are the total others for that neoadjuvant treatment cohort (e.g., in the overall cohort, 145 patients had ‘other’).

    EBRT: External beam radiation therapy; IMRT: Intensity-modulated radiation therapy; SBRT/SABR: Stereotactic body radiation therapy/stereotactic ablative radiotherapy; SD: Standard deviation.

    Among the patients treated with neoadjuvant chemoradiotherapy, the median (range) duration of treatment was 6.3 (1.0, 32.9) weeks or 3 (1, 16) cycles (Table 2). All except for two patients (docetaxel and paclitaxel monotherapies) received platinum-based therapies; 59.0% received carboplatin-based and 40.3% received cisplatin-based. The most common neoadjuvant regimens were paclitaxel + carboplatin (56.8%), cisplatin + etoposide (35.9%), docetaxel + cisplatin (9.5%) and gemcitabine + cisplatin (9.5%). Nearly all (95%) patients received radiation therapy concurrent with neoadjuvant chemotherapy. The median (range) dose was 8.4 Gy (0.67, 71.20).

    Demographics & clinical characteristics by surgery status

    A total of 225 (52.9%) patients in the overall study population underwent a surgical resection. Patients who underwent surgical resection were younger, with a median (range) age of 64 years (27, 89) versus 68 years (41, 87). They were also less likely to be current or former smokers (64.0 vs 72.5%) or have an ECOG PS score of 1 or 2 (60.8 vs 72.5%). Patients who underwent surgical resection included a higher proportion who had nonsquamous histology (57.3 vs 46.5%).

    Among the patients who did not undergo surgical resection (n = 200), the most common reasons (among ≥5% of those patients) were physician recommendation (n = 72, 36.0%), disease progression (n = 55, 27.5%), patient preference (n = 14, 7.0%) and death (n = 14, 7.0%).

    Pathologic complete response

    Of the 425 patients in the overall population, 48 (11.3%) patients achieved a pCR. The pCR rates among patients initiated on neoadjuvant chemotherapy and chemoradiotherapy were 4.8 and 14.7%, respectively. In the overall population, 184 patients (43.3% of overall) died during the study observation period: 16 (33.3%) and 168 (44.6%) among patients with and without pCR, respectively. The median OS (95% CI) among patients with a pCR was 92.9 months (60.5–125.1) compared with 41.0 months (30.6–53.6) among patients without a pCR (Figure 2A). A total of 264 (62.1%) patients had an EFS event (progression/death) during the study observation period with 22 (45.8%) and 242 (64.2%) among patients with and without pCR, respectively. The median (95% CI) EFS among patients with a pCR was 64.5 months (25.6-NR) compared with 13.0 months (10.8–15.2) among patients without a pCR (Figure 2B). Compared with no pCR, pCR was shown to be associated with better OS and EFS by univariate and multivariable Cox analysis (Table 3). In subgroup analyses, pCR was associated with OS in patients with stage III (HR = 0.46, 95% CI: 0.22–0.95 in patients with pCR vs not) and with EFS in patients with stages I/II and III (HR = 0.43, 95% CI: 0.24–0.78 in patients with stage III, HR = 0.51, 95% CI: 0.28–0.94 in patients with stages I/II). However, in patients treated with chemotherapy, the risk of an event was numerically lower in patients who achieved a pCR versus those who did not, but this was not statistically significant (HR = 0.22, 95% CI: 0.03–1.59 for OS, HR = 0.26, 95% CI: 0.06–1.05 for EFS; data not shown). In a sensitivity analysis of only patients who underwent resection, the association between EFS and pCR remained significant (HR = 0.61; 95% CI: 0.38–0.98) in the multivariable analysis, but the association between OS and pCR was not as strong in the univariate analysis (HR = 0.81; 95% CI: 0.47–1.41).

    Figure 2. Kaplan-Meier curves for survival outcomes by response status for the overall study population.

    (A) Overall survival by pathologic complete response status, (B) event-free survival by pathologic complete response status, (C) overall survival by major pathologic response status and (D) event-free survival by major pathologic response status.

    HR: Hazard ratio; NR: Not reached; pCR: Pathologic complete response.

    Table 3. Association of pathologic complete response and other covariates with overall survival and event-free survival, Cox analysis (overall study population).
    CovariateLevelOS univariate Cox analysisOS multivariable Cox analysisEFS univariate Cox analysisEFS multivariable Cox analysis
      Unadjusted hazard ratios (95% CI)p-valueAdjusted hazard ratios (95% CI)p-valueUnadjusted hazard ratios (95% CI)p-valueAdjusted hazard ratios (95% CI)p-value
    AgeContinuous1.01 (0.99–1.03)0.161.01 (0.99–1.03)0.131.01 (0.99–1.02)0.4177  
    Age<45 yearsReferent0.623  Referent0.5617  
    45–54 years1.03 (0.38–2.81)   1.44 (0.63–3.28)   
    55–64 years1.22 (0.49–3.05)   1.26 (0.58–2.72)   
    65–74 years1.32 (0.53–3.28)   1.46 (0.68–3.15)   
    75+ years1.56 (0.61–3.99)   1.62 (0.73–3.59)   
    GenderMaleReferent0.11 0.11Referent0.261  
    Female0.79 (0.59–1.06) 0.79 (0.59–1.06) 0.87 (0.68–1.11)   
    RaceCaucasianReferent<0.0001 <0.0001Referent<0.0001 <0.0001
    African American1.04 (0.51–2.12) 0.92 (0.45–1.88) 1.99 (1.21–3.27) 1.79 (1.09–2.96) 
    Suppressed0.75 (0.24–2.36) 0.57 (0.18–1.83) 0.96 (0.39–2.34) 0.84 (0.34–2.06) 
    Other§0.71 (0.33–1.53) 0.64 (0.30–1.39) 0.96 (0.55–1.69) 0.89 (0.50–1.57) 
    Not documented2.89 (2.04–4.08) 2.91 (2.05–4.14) 2.01 (1.49–2.72) 2.07 (1.53–2.81) 
    HistologyNon-squamousReferent0.22  Referent0.8879  
    Squamous1.23 (0.91–1.66)   0.96 (0.75–1.24)   
    No info1.49 (0.86–2.58)   1.08 (0.66–1.77)   
    Initial tumor stageI & IIReferent0.06 0.29Referent0.0465 0.7942
    III1.35 (0.99–1.85) 1.19 (0.86–1.66) 1.30 (1.00–1.69) 1.04 (0.79–1.35) 
    Neoadjuvant therapyChemotherapyReferent0.08 0.18Referent0.0253 0.0928
    Chemoradiotherapy0.76 (0.56–1.03) 0.81 (0.59–1.10) 0.75 (0.58–0.97) 0.80 (0.622–1.04) 
    pCRNo pCRReferent0.006 0.01Referent0.0001 0.0003
    pCR0.49 (0.29–0.81) 0.50 (0.30–0.85) 0.43 (0.27–0.66) 0.44 (0.28–0.68) 

    †The variables with a p-value less than or equal to 0.20 in Cox univariate analyses were included in the multivariable analyses.

    ‡Suppressed: information was not reported for variables with n≤5 for HIPAA reasons.

    §Other included Asian race, declined to specify, other (not described further for HIPAA reasons for variables with n≤5) and unknown.

    EFS: Event-free survival; HIPAA: Health Insurance Portability and Accountability Act of 1996; OS: Overall survival; pCR: Pathologic complete response.

    Major pathologic response

    Of the 425 patients in the overall population, 80 (18.8%) patients achieved an MPR. The MPR rates among patients initiated on neoadjuvant chemotherapy and chemoradiotherapy were 8.8 and 24.1%, respectively. The median OS (95% CI) among patients with an MPR (total n = 80; 25 events, 31.3%) was 92.9 months (59.0–125.1) compared with 37.3 months (28.2–52.0) among patients without an MPR (total n = 345; 159 events, 46.1%; Figure 2C). The median EFS (95% CI) among patients with a MPR (total n = 80; 39 events, 48.8%) was 37.8 (25.2–NR) months compared with 12.9 (9.9–14.4) months among patients without MPR (total n = 345; 225 events, 65.2%; Figure 2D). MPR was shown to be associated with better OS and EFS by univariate and multivariable Cox analysis (Table 4). Among patients treated with chemoradiotherapy compared with chemotherapy as the reference, MPR was also associated with better OS (HR = 0.48; 95% CI 0.30–0.77; p = 0.0024) and EFS (HR = 0.506; 95% CI 0.347–0.738; p = 0.0004). Among patients who had undergone resection, in a univariate Cox analysis, MPR did not have a significant association with OS (HR = 0.84; 95% CI: 0.52–1.37; p = 0.4908) or with EFS (HR = 0.76; 95% CI: 0.51–1.11; p = 0.1527). In subgroup analyses, MPR in stages I/II and III was associated with OS (HR = 0.42, 95% CI: 0.21–0.86 and HR = 0.52, 95% CI: 0.30–0.89, respectively) and EFS (HR = 0.46, 95% CI: 0.27–0.80 and HR = 0.48, 95% CI: 0.31–0.74, respectively), and although the risk for an event was numerically lower for patients treated with chemotherapy who achieved an MPR compared with patients who did not achieve MPR, it was only statistically significant for EFS (HR = 0.59, 95% CI: 0.22–1.63 for OS; HR = 0.37, 95% CI: 0.15–0.90 for EFS; data not shown).

    Table 4. Association of major pathologic response and other variables with overall survival and event-free survival, Cox analysis (overall study population).
    CovariateLevelOS univariate cox analysisOS multivariable cox analysisEFS univariate cox analysisEFS multivariable cox analysis
      Unadjusted hazard ratios (95% CI)p-valueAdjusted hazard ratios (95% CI)p-valueUnadjusted hazard ratios (95% CI)p-valueAdjusted hazard ratios (95% CI)p-value
    AgeContinuous1.01 (0.99-1.03)0.15671.01 (0.99-1.03)0.14841.01 (0.99–1.02)0.4177  
    Age<45 yearsReferent0.6433  Referent0.5617  
    45–54 years0.98 (0.36-2.66)   1.44 (0.63–3.28)   
    55–64 years1.17 (0.47-2.93)   1.26 (0.58–2.72)   
    65–74 years1.29 (0.52-3.19)   1.46 (0.68–3.15)   
    75+ years1.48 (0.58-3.78)   1.62 (0.73–3.59)   
    GenderMaleReferent0.0721  Referent0.261  
    Female0.76 (0.57-1.02) 0.72 (0.54-0.97)0.03140.87 (0.68–1.11)   
    RaceCaucasianReferent<0.0001  Referent<0.0001 <0.0001
    African American1.02 (0.49-2.09) 0.95 (0.46-1.95)<0.00011.99 (1.21–3.27) 1.85 (1.12–3.06) 
    Suppressed0.77 (0.24-2.42) 0.61 (0.19-1.96) 0.96 (0.39–2.34) 0.89 (0.36–2.17) 
    Other§0.71 (0.33-1.53) 0.61 (0.28-1.33) 0.96 (0.55–1.69) 0.84 (0.48–1.49) 
    Not documented2.45 (1.75-3.45) 0.61 (0.28-1.33) 2.01 (1.49–2.72) 1.99 (1.47–2.71) 
    HistologyNon-SquamousReferent0.3642  Referent0.8879  
    Squamous1.22 (0.89-1.65)   0.96 (0.75–1.24)   
    No info1.31 (0.76-2.26)   1.08 (0.66–1.77)   
    Initial tumor stage I & IIReferent0.0931  Referent0.0764 0.8005
    III1.31 (0.96-1.79) 1.14 (0.82-1.59)0.43121.27 (0.98–1.64) 1.04 (0.79–1.35) 
    Neoadjuvant therapy ChemotherapyReferent0.1132  Referent0.0253 0.1465
    Chemoradiotherapy0.78 (0.58-1.06) 0.86 (0.63-1.18)0.35420.75 (0.58–0.97) 0.83 (0.64–1.07) 
    MPR No MPRReferent0.0008  Referent<0.0001 <0.0001
    MPR0.49 (0.32-0.74) 0.52 (0.34-0.81)0.00340.47 (0.33–0.66) 0.49 (0.34–0.69) 

    †The variables with a p-value less than or equal to 0.20 in Cox univariate analyses were included in the multivariable analyses.

    ‡Information not reported for HIPAA reasons for variables with n≤5.

    §Other included Asian race, declined to specify, other (not described further for HIPAA reasons for variables with n≤5) and unknown.

    EFS: Event-free survival; HIPAA: Health Insurance Portability and Accountability Act of 1996; MPR: Major pathologic response; OS: Overall survival.

    EFS & OS correlation

    Landmark analysis/time-dependent analysis

    In landmark analyses at the different time points (6, 12 and 24 months), patients without an EFS event were shown to have significantly better OS than those with an EFS event (HR = 0.34; 95% CI: 0.23–0.50; HR = 0.24; 95% CI: 0.16–0.36; and HR = 0.29; 95% CI: 0.17–0.50, respectively; Figure 3A, B & C respectively). This was also evident in a Cox analysis where the event was considered as a time dependent variable (unadjusted HR = 0.51; 95% CI: 0.38–0.59; Supplementary Table 1).

    Figure 3. Landmark overall survival Kaplan-Meier curve by the presence of event-free survival event.

    (A) 6 months, (B) 12 months and (C) 24 months.

    EFS: Event-free survival; HR: Hazard ratio; NR: Not reached.

    Correlation analysis

    The correlation between EFS and OS was tested using the Kendall's Tau rank correlation method. Considering all patients who initiated neoadjuvant therapy, the correlation between EFS and OS was 0.66 (p < 0.001) in all patients (n = 425) and 0.72 (p < 0.001) for patients who underwent a surgical resection (n = 225). As there was no established threshold for interpreting Kendall's Tau (that we are aware of), we noted that EFS was moderately correlated with OS according to Chan [26] (Supplementary Figure 1).

    Discussion

    In this real-world study of 425 patients with resectable NSCLC treated with neoadjuvant chemotherapy or chemoradiotherapy in USA, achieving pCR or MPR was associated with significantly longer survival. More specifically, among the overall population, patients who achieved a pCR or MPR had approximately half the mortality risk compared with those who did not achieve a pCR or MPR.

    These results were in line with large meta-analyses that found an association between pathologic response and survival in patients receiving neoadjuvant chemotherapy or chemoradiotherapy, increasing the confidence of a true association between pCR and longer-term efficacy end points [19,20]. The association between pCR and EFS was also observed in the CheckMate 816 trial, a phase III clinical trial comparing neoadjuvant immunotherapy (nivolumab + chemotherapy vs chemotherapy alone) where patients who experienced a pCR had longer EFS compared with those without a pCR (HR = 0.13, 95% CI: 0.05–0.37) [27,28].

    Our study populations were generally similar to the studies included in the meta-analysis (resectable NSCLC treated with neoadjuvant chemotherapy ± radiotherapy). However, caution should be exercised when comparing the two, given the differing data sources (real-world study vs literature review including various study designs such as cohorts, single-arm trials and randomized controlled trials [RCT]). It is encouraging to see similar trends of improved survival outcomes for the patients with NSCLC who achieved a pathologic response (relative to those who did not) after neoadjuvant treatment from a literature review, after neoadjuvant immunotherapy in a phase III RCT, and now in a real-world study. Although all patients had the intention to undergo surgery, approximately 50% of the patients who were initiated on neoadjuvant therapy did not undergo surgery in this real-world study. Physician recommendation, disease progression and patient preference were the most common reasons for not proceeding with surgery among these patients.

    In this study, the landmark analysis considering EFS events at different landmark points showed a decreased risk for death among patients who did not have an EFS event. The landmark method, which avoids the immortal time bias in the estimation of the OS by re-baselining patients and accounting for outcomes at a landmark time point, has been proposed as an appropriate method to estimate the survival based on the responses at the different points in time [30–34]. When utilizing a time-dependent Cox analysis, we found that EFS was moderately correlated with OS (according to Chan) [26]. Given the attenuation bias because of censoring of survival, the results of the correlation may be underestimated due to lack of visibility of events occurring after the follow-up period. A meta-analysis of patients treated with neoadjuvant therapy for resectable NSCLC also found a correlation between EFS with OS [35].

    Acknowledging the time bias carried by analyses of survival by response, in the context of surrogacy and association of end points, these summaries of EFS/OS by pCR/MPR status ensure that the full EFS/OS definition information (including events occurring before surgery) is used in the assessment, including early events. Due to the limited size of this study (N = 425), it is challenging to reliably explore the various clinical subgroups of interest, particularly in the context of a limited number of patients achieving pCR with chemotherapy/chemoradiotherapy. In the sensitivity analysis of only patients undergoing surgical resection, the magnitude of the association between pCR/MPR and EFS/OS was not as strong as in the overall population, though the HR point estimates for EFS and OS were both <1. These results should be interpreted with caution, as the sample size was small and selection of those patients was biased by events occurring after the index date.

    Of note, the study identification period for this real-world study spans over 10 years, during which therapeutic options for management of advanced NSCLC have evolved significantly. The use of staging methods such as PET/CT scanning and endobronchial ultrasound biopsy might have changed [36,37]. Immunotherapy and targeted therapy have changed the landscape of lung cancer management, leading to improved survival rates. Further, data from phase III studies in populations with non-metastatic, resectable NSCLC are still emerging in the neoadjuvant and adjuvant settings [27,38]. The associations between early end points and survival in this study are based on chemotherapy- and chemoradiotherapy-based neoadjuvant treatments, which have shown only modest survival benefit versus no treatment in this setting (∼5% OS absolute risk reduction at 5 years) [10]. With newer advances such as immunotherapy, larger increases in pCR rates in RCTs are expected. When more trial data become available in patients treated with immunotherapy in the neoadjuvant setting, it will be useful to conduct a meta-analysis to show whether a trial-level association holds true as well in this patient population.

    Strengths/limitations

    This study has several strengths to be highlighted. The study investigated a large representative population of patients with NSCLC receiving care in the US community oncology setting. The iKM data include the documentation of diagnostic, clinical, treatment characteristics and outcomes data outside clinical trials. Therefore, the study could capture the real-world experiences of patients with cancer with up to 10 years of follow-up. The most widely used definition of pCR (0% viable tumor in the primary tumor and nodes) was used in this study to minimize definition heterogeneity in the primary analyses. Different networks may follow different treatment protocols. However, The US Oncology Network utilizes National Comprehensive Cancer Network guidelines, which is a common standard across the USA.

    As with any database study, there are limitations to be considered when interpreting data analyzed from an EHR database. While the iKM database contains a wealth of information about community-based oncology, it may be limited in various aspects. First, the iKM system is used for clinical practice purposes, not for research purposes. As such, associations but not causality can be detected. Second, not all community oncology practices are included in the iKM dataset. Furthermore, not all The US Oncology Network clinics utilize the full EHR/iKM capabilities. Those practices that participate in the Network may be different from those that do not participate in some way, such as the patient population that is seen or the prescribing practices of the physicians. Third, the EHR data do not capture information about services and procedures provided outside of the practice if not documented in the EHR; therefore, results may not be generalized to the US population without further evaluation. Some variables of interest may not be as complete across the entire population.

    Although surgery was planned in all patients receiving neoadjuvant treatment, approximately 50% of the patients did not eventually undergo surgery, thus reducing the number of patients with available data for study end points.

    As these are real-world data, potential heterogeneity in pathologic definitions and analyses between pathologists and study sites may exist. Particularly for this study, it was important to standardize the definition of pCR to the highest degree possible across the study population. The study defines pCR as ypT0N0, which was the highest available standard based on the data collected, and included all treated patients with resectable intent, so to avoid biasing the study with events occurring after the index date. The pathologists who provide services to The US Oncology Network follow the College of American Pathologists guidelines for consistent identification and reporting of cancer, which acts as a standardization process. However, comprehensive data on residual tumor were not available for the lymph nodes and hence, the definition of MPR had to be modified from what was included in the study protocol. Also, while pathology reports were used to determine pathologic response rate, the scoring methods used for assessment were not provided.

    Conclusion

    In this real-world study among patients initiating chemotherapy or chemoradiotherapy as neoadjuvant therapy with an intention of surgery, achieving pCR or MPR was associated with significantly better OS and EFS compared with those patients without a pCR or MPR. In addition, patients with longer EFS had longer OS. These results suggest that pathologic response and EFS may have utility as candidate surrogate end points, which would have great impact on allowing clinicians to evaluate early response to a treatment and on shortening drug development timelines to get effective drugs to patients faster.

    Summary points
    • We examined how early end points (pathologic response) and long-term end points (survival) are related in patients with resectable non-small-cell lung cancer in a large network of community-based oncology practices.

    • Patients with complete pathologic response or major pathologic response had better overall and event-free survival than those without.

    • Longer event-free survival was associated with longer overall survival.

    • This study provides real-world evidence to support the data that pathologic response and event-free survival may be useful early surrogates for longer-term survival end points.

    • Use of early end points would allow assessment of early response to treatment, which can allow clinicians to evaluate early response to a treatment and could shorten new drug development timelines.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/fon-2023-0170

    Acknowledgments

    L Kaspin-Powell provided medical writing and editorial assistance.

    Financial & competing interests disclosure

    This study was funded by Bristol Myers Squibb. E Nadler has received payment for speaker bureau and speaker arrangement from AstraZeneca, Genentech, G1 Therapeutics, Merck and Regeneron. A Vasudevan was an employee of Ontada at the time the study was performed. C Wentworth and Nicholas Robert are employees of Ontada. J Fiore, JR Penrod and L Vo are employees of Bristol Myers Squibb. J Fiore and JR Penrod also hold stock in Bristol-Myers Squibb. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    L Kaspin-Powell provided medical writing and editorial assistance.

    Ethical conduct of research

    Institutional Review Board and Compliance/Privacy approval was gained prior to initiation of the retrospective research. Since this project involved the analysis of existing data and records, study information was analyzed in such a manner that research participants could not be directly identified. Patient informed consent was not required due to the nature of the study design. Thus, exemption status and a waiver of informed consent were approved by The US Oncology, Inc. Institutional Review Board. Data were handled in compliance with HIPAA and the Health Information Technology for Economic and Clinical Health (HITECH) Act.

    Availability of data & material

    The raw data used for this analysis are not publicly available due to privacy or ethical restrictions.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Morgensztern D, Ng SH, Gao F, Govindan R. Trends in stage distribution for patients with non-small-cell lung cancer: a National Cancer Database survey. J. Thorac. Oncol. 5(1), 29–33 (2010).
    • 2. Uramoto H, Tanaka F. Recurrence after surgery in patients with NSCLC. Transl. Lung Cancer Res. 3(4), 242–249 (2014).
    • 3. al-Kattan K, Sepsas E, Fountain SW, Townsend ER. Disease recurrence after resection for stage I lung cancer. Eur. J. Cardiothorac. Surg. 12(3), 380–384 (1997).
    • 4. Su S, Scott WJ, Allen MS et al. Patterns of survival and recurrence after surgical treatment of early stage non-small-cell lung carcinoma in the ACOSOG Z0030 (ALLIANCE) trial. J. Thorac. Cardiovasc. Surg. 147(2), 747–752; discussion 752–743 (2014).
    • 5. Martini N, Bains MS, Burt ME et al. Incidence of local recurrence and second primary tumors in resected stage I lung cancer. J. Thorac. Cardiovasc. Surg. 109(1), 120–129 (1995).
    • 6. Fedor D, Johnson WR, Singhal S. Local recurrence following lung cancer surgery: incidence, risk factors, and outcomes. Surg. Oncol. 22(3), 156–161 (2013).
    • 7. NSCLC Meta-analysis Collaborative Group. Preoperative chemotherapy for non-small-cell lung cancer: a systematic review and meta-analysis of individual participant data. Lancet 383(9928), 1561–1571 (2014).
    • 8. Ettinger DS, Wood DE, Akerley W et al. Non-small-cell lung cancer, version 1.2015. J. Natl Compr. Canc. Netw. 12(12), 1738–1761 (2014).
    • 9. Ettinger DS, Akerley W, Bepler G et al. Non-small-cell lung cancer. J. Natl Compr. Canc. Netw. 8(7), 740–801 (2010).
    • 10. Lim E, Harris G, Patel A, Adachi I, Edmonds L, Song F. Preoperative versus postoperative chemotherapy in patients with resectable non-small-cell lung cancer: systematic review and indirect comparison meta-analysis of randomized trials. J. Thorac. Oncol. 4(11), 1380–1388 (2009).
    • 11. Pisters KM, Le Chevalier T. Adjuvant chemotherapy in completely resected non-small-cell lung cancer. J. Clin. Oncol. 23(14), 3270–3278 (2005).
    • 12. Rosell R, Gómez-Codina J, Camps C et al. A randomized trial comparing preoperative chemotherapy plus surgery with surgery alone in patients with non-small-cell lung cancer. N. Engl. J. Med. 330(3), 153–158 (1994).
    • 13. Roth JA, Fossella F, Komaki R et al. A randomized trial comparing perioperative chemotherapy and surgery with surgery alone in resectable stage IIIA non-small-cell lung cancer. J. Natl Cancer Inst. 86(9), 673–680 (1994).
    • 14. Arriagada R, Auperin A, Burdett S et al. Adjuvant chemotherapy, with or without postoperative radiotherapy, in operable non-small-cell lung cancer: two meta-analyses of individual patient data. Lancet 375(9722), 1267–1277 (2010).
    • 15. Brahmer J, Reckamp KL, Baas P et al. Nivolumab versus Docetaxel in advanced squamous-cell non-small-cell lung cancer. N. Engl. J. Med. 373(2), 123–135 (2015).
    • 16. Provencio M, Nadal E, Insa A et al. Neoadjuvant chemo-immunotherapy for the treatment of stage IIIA resectable non-small-cell lung cancer (NSCLC): a Phase II multicenter exploratory study—Final data of patients who underwent surgical assessment. J. Clin. Oncol. 37(Suppl. 15), 8509 (2019).
    • 17. Hellmann MD, Chaft JE, William WN Jr et al. Pathological response after neoadjuvant chemotherapy in resectable non-small-cell lung cancers: proposal for the use of major pathological response as a surrogate end point. Lancet Oncol. 15(1), e42–50 (2014). •• This paper makes the case for the use of major pathological response as a surrogate for overall survival in clinical trials of new cancer drugs
    • 18. Clinical Trial End points for the Approval of Non Small Cell Lung Cancer. Drugs and Biologics Guidance for Industry. US Department of Health and Human Services, US Food and Drug Administration, Center for Drug Evaluation and Research, Center for Biologics Evaluation and Research (2015). https://www.fda.gov/media/116860/download (Accessed 6 September 2022). • This US FDA document mentions how the accelerated approval pathway can use surrogate end points that can be measured earlier and still predict clinical benefit.
    • 19. Waser NA, Adam A, Schweikert B et al. Pathologic response as early end point for survival following neo-adjuvant therapy (NEO-AT) in resectable non-small-cell lung cancer (rNSCLC): systematic literature review and meta-analysis (Abstract 1243P). Ann. Oncol. 31(Suppl. 4), S806 (2020). •• This systematic review and meta-analysis found a strong association between pathological response and survival in patients with non-small-cell lung cancer
    • 20. Rosner S, Liu C, Forde PM, Hu C. Association of pathologic complete response and long-term survival outcomes among patients treated with neoadjuvant chemotherapy or chemoradiotherapy for NSCLC: a meta-analysis. JTO Clin Res Rep. 3(9), 100384 (2022). •• This systematic review and meta-analysis found a strong association between pathological response and survival (both event-free and overall) in patients with non-small-cell lung cancer.
    • 21. Blumenthal GM, Bunn PA Jr, Chaft JE et al. Current status and future perspectives on neoadjuvant therapy in lung cancer. J. Thorac. Oncol. 13(12), 1818–1831 (2018).
    • 22. Cottrell TR, Thompson ED, Forde PM et al. Pathologic features of response to neoadjuvant anti-PD-1 in resected non-small-cell lung carcinoma: a proposal for quantitative immune-related pathologic response criteria (irPRC). Ann. Oncol. 29(8), 1853–1860 (2018).
    • 23. American Joint Committee on Cancer. AJCC Cancer Staging Manual. Seventh Edition (2010). www.facs.org/media/j30havyf/ajcc_7thed_cancer_staging_manual.pdf (Accessed 1 December 2022).
    • 24. Rami-Porta R, Asamura H, Travis WD, Rusch VW. Lung cancer - major changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J. Clin. 67(2), 138–155 (2017).
    • 25. The US Oncology Network. https://www.usoncology.com/our-company
    • 26. Chan YH. Biostatistics 104: correlational analysis. Singapore Med. J. 44(12), 614–619 (2003).
    • 27. Forde PM, Spicer J, Lu S et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N. Engl. J. Med. 386(21), 1973–1985 (2022).
    • 28. Provencio-Pulla M, Spicer J, Taube JM et al. Neoadjuvant nivolumab (NIVO) + platinum-doublet chemotherapy (chemo) versus chemo for resectable (IB–IIIA) non-small-cell lung cancer (NSCLC): association of pathological regression with event-free survival (EFS) in CheckMate 816. J. Clin. Oncol. 40(Suppl. 17), LBA8511 (2022). •• This paper reports on a post-hoc analysis of a clinical trial that had previously found improvement in both event-free survival and pathologic complete response; the post-hoc analysis found an association between event-free survival and pathologic complete response.
    • 29. Kalhor N, Correa AM. Correa AM et al. Histopathologic response criteria predict survival of patients with resected lung cancer after neoadjuvant chemotherapy. J Thorac Oncol. 7(5), 825–832 (2012).
    • 30. Anderson JR, Cain KC, Gelber RD. Analysis of survival by tumor response. J. Clin. Oncol. 1(11), 710–719 (1983).
    • 31. Antonia SJ, Borghaei H, Ramalingam SS et al. Four-year survival with nivolumab in patients with previously treated advanced non-small-cell lung cancer: a pooled analysis. Lancet Oncol. 20(10), 1395–1408 (2019).
    • 32. Hellmann MD, Ciuleanu TE, Pluzanski A et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N. Engl. J. Med. 378(22), 2093–2104 (2018).
    • 33. Hellmann MD, Paz-Ares L, Bernabe Caro R et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N. Engl. J. Med. 381(21), 2020–2031 (2019).
    • 34. Santi I, Smare C, Juarez-Garcia A et al. PRM157 - The Impact of Landmark Response on Overall Survival: Implications for the Economic Evaluation of Immune-Therapy (I-O) Treatment in Small Cell Lung Cancer (SCLC). Value in Health. 21, S383 (2018).
    • 35. Ostoros G, Berktas M, Chander P et al. Association between event-free survival and overall survival following neoadjuvant therapy for resectable non-small-cell lung cancer. Presented at: European Society for Medical Oncology Congress. Paris, France, Poster Session 943P, 9–13 September 2022. (Accessed 3 October 2022). https://oncologypro.esmo.org/meeting-resources/esmo-congress/association-between-event-free-survival-and-overall-survival-following-neoadjuvant-therapy-for-non-small-cell-lung-cancer •• This systematic review and meta-analysis found a strong association between pathological response and survival (both event-free and overall) in patients with non-small-cell lung cancer
    • 36. Counts SJ, Kim AW. Diagnostic imaging and newer modalities for thoracic diseases: PET/computed tomographic imaging and endobronchial ultrasound for staging and its implication for lung cancer. Surg. Clin. North Am. 97(4), 733–750 (2017).
    • 37. Rami-Porta R, Call S, Dooms C et al. Lung cancer staging: a concise update. Eur. Respir. J. 51(5), 1800190 (2018).
    • 38. Felip E, Altorki N, Zhou C et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, Phase III trial. Lancet 398(10308), 1344–1357 (2021).

    1 Diagnosed prior to January 2018, and therefore was based on TNM 7th edition.

    2 Diagnosed on or after January 2018, and therefore based on TNM 8th edition.