Disseminated Tumor Cells in Lymph Nodes as a Determinant for Survival in Surgically Resected Non–Small-Cell Lung Cancer

1999 ◽  
Vol 17 (1) ◽  
pp. 19-19 ◽  
Author(s):  
B. Kubuschok ◽  
B. Passlick ◽  
J. R. Izbicki ◽  
O. Thetter ◽  
K. Pantel

PURPOSE: In recent years, the detection of even a few tumor cells in lymph nodes of patients with surgically resected non–small-cell lung cancer (NSCLC) became possible with immunohistochemical staining procedures. Tumor cells in lymph nodes have been shown to be associated with an increased rate of early recurrence. However, the prognostic significance of this minimal tumor cell spread for overall survival remains unclear. PATIENTS AND METHODS: We used the epithelium-specific monoclonal antibody Ber-EP4, which recognizes the 17-1A antigen (also called EGP40 or Ep-CAM), to discover small tumor cell deposits (≤ three cells) in 565 regional lymph nodes judged as tumor-free by conventional histopathology in patients with NSCLC staged as pT1-4, pN0-2, M0, R0. In a prospective analysis, we studied the influence of the detected tumor cells on the cancer recurrence rate and survival of 117 patients. RESULTS: Ber-EP4-positive cells were found in 27 of 125 patients (21.6%). After an observation period of 64 months, patients with disseminated tumor cells had reduced disease-free survival (P < .0001) and overall survival (P = .0001) rates in univariate analyses (log-rank test). Multivariate analysis (Cox model) showed a 2.7 times increased risk for tumor relapse and a 2.5 times increased risk for shorter survival in patients with disseminated tumor cells compared with patients without such cells. Patients without any evidence of histopathologic and immunohistochemical lymph node involvement had an overall survival rate of 78%. CONCLUSION: The immunohistochemical detection of disseminated tumor cells in lymph nodes of patients with completely resected NSCLC is an independent prognostic factor for overall survival.

2016 ◽  
Vol 11 (1) ◽  
Author(s):  
Ane Kongsgaard Rud ◽  
Kjetil Boye ◽  
Øystein Fodstad ◽  
Siri Juell ◽  
Lars H. Jørgensen ◽  
...  

Lung Cancer ◽  
1997 ◽  
Vol 18 ◽  
pp. 229-230
Author(s):  
U. Seifart ◽  
S. Henrich ◽  
G. Jaques ◽  
C. Loechelt ◽  
A. Wachtel ◽  
...  

2020 ◽  
Vol 15 (1) ◽  
Author(s):  
Kathrin Gennen ◽  
Lukas Käsmann ◽  
Julian Taugner ◽  
Chukwuka Eze ◽  
Monika Karin ◽  
...  

Abstract Background/aim mmune checkpoint inhibition (CPI) has an increasing impact in the multimodal treatment of locally advanced non-small cell lung cancer (LA-NSCLC). Increasing evidence suggests treatment outcome depending on tumor cell PD-L1 expression. The purpose of this retrospective study was to investigate the prognostic value of PD-L1 expression on tumor cells in combination with CD8+ tumor stroma-infiltrating lymphocyte (TIL) density in inoperable LA-NSCLC treated with concurrent chemoradiotherapy (CRT). Patients and method We retrospectively assessed clinical characteristics and initial tumor biopsy samples of 31 inoperable LA-NSCLC patients treated with concurrent CRT. Prognostic impact of tumor cell PD-L1 expression (0% versus ≥1%) and CD8+ TIL density (0–40% vs. 41–100%) for local control, progression-free (PFS) and overall survival (OS) as well as correlations with clinicopathological features were evaluated. Results Median OS was 14 months (range: 3–167 months). The OS rates at 1- and 2 years were 68 and 20%. Local control of the entire cohort at 1 and 2 years were 74 and 61%. Median PFS, 1-year and 2-year PFS were 13 ± 1.4 months, 58 and 19%. PD-L1 expression < 1% on tumor cells was associated with improved OS, PFS and local control in patients treated with concurrent CRT. Univariate analysis showed a trend towards improved OS and local control in patients with low CD8+ TIL density. Evaluation of Tumor Immunity in the MicroEnvironment (TIME) appears to be an independent prognostic factor for local control, PFS and OS. The longest and shortest OS were achieved in patients with type I (PD-L1neg/CD8low) and type IV (PD-L1pos/CD8low) tumors (median OS: 57 ± 37 vs. 10 ± 5 months, p = 0.05), respectively. Conclusion Assessment of PD-L1 expression on tumor cells in combination with CD8+ TIL density can be a predictive biomarker in patients with inoperable LA-NSCLC treated with concurrent CRT.


Cells ◽  
2019 ◽  
Vol 8 (8) ◽  
pp. 880 ◽  
Author(s):  
Eva Obermayr ◽  
Christiane Agreiter ◽  
Eva Schuster ◽  
Hannah Fabikan ◽  
Christoph Weinlinger ◽  
...  

At initial diagnosis, most patients with small-cell lung cancer (SCLC) present with metastatic disease with a high number of tumor cells (CTCs) circulating in the blood. We analyzed RNA transcripts specific for neuroendocrine and for epithelial cell lineages, and Notch pathway delta-like 3 ligand (DLL3), the actionable target of rovalpituzumab tesirine (Rova-T) in CTC samples. Peripheral blood samples from 48 SCLC patients were processed using the microfluidic Parsortix™ technology to enrich the CTCs. Blood samples from 26 healthy donors processed in the same way served as negative controls. The isolated cells were analyzed for the presence of above-mentioned transcripts using quantitative PCR. In total, 16/51 (31.4%) samples were CTC-positive as determined by the expression of epithelial cell adhesion molecule 1 (EpCAM), cytokeratin 19 (CK19), chromogranin A (CHGA), and/or synaptophysis (SYP). The epithelial cell lineage-specific EpCAM and/or CK19 gene expression was observed in 11 (21.6%) samples, and positivity was not associated with impaired survival. The neuroendocrine cell lineage-specific CHGA and/or SYP were positive in 13 (25.5%) samples, and positivity was associated with poor overall survival. DLL3 transcripts were observed in four (7.8%) SCLC blood samples and DLL3-positivity was similarly associated with poor overall survival (OS). CTCs in SCLC patients can be assessed using epithelial and neuroendocrine cell lineage markers at the molecular level. Thus, the implementation of liquid biopsy may improve the management of lung cancer patients, in terms of a faster diagnosis, patient stratification, and on-treatment therapy monitoring.


2016 ◽  
Vol 34 (15_suppl) ◽  
pp. 8547-8547
Author(s):  
Ahmedin Jemal ◽  
Chun Chieh Lin ◽  
Matthew Smeltzer ◽  
Raymond U. Osarogiagbon

2012 ◽  
Vol 7 (7) ◽  
pp. 1202-1203
Author(s):  
Anna Puggina ◽  
Verena Kümmerlen ◽  
Korinna Jöhrens-Leder ◽  
Ulrich Keilholz ◽  
Alberto Fusi

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A392-A393
Author(s):  
Guenter Schmidt ◽  
Ansh Kapil ◽  
Lina Meinecke ◽  
Farzad Sekhavati ◽  
Jan Lesniak ◽  
...  

BackgroundThe pathologist’s visual assessment of tumor proportion score (TPS) with 25% cutoff on PD-L1 stained tissue samples is an established method to select metastatic NSCLC patients that are likely to respond to an anti-PD-L1 monotherapy.1 However, manual scoring is often subject to subjectivity in human perception2 and there remains a critical need for more objective and quantitative methods to assess PD-L1 expression in immuno-oncology.MethodsWe used deep learning (DL) based image analysis (IA) to generate a novel PD-L1 Quantitative Continuous Score (QCS)3 in tumor cells. PD-L1 QCS consists of two DL models to first segment epithelial regions and second detect membranes, cytoplasm and nuclei of each tumor cell in PD-L1 immunohistochemically (IHC) stained tissue slides. The PD-L1 expression of each tumor cell compartment was estimated by the respective optical density (OD) of DAB, and tumor cells with a membrane OD greater than ODmin are considered as PD-L1-positive. A slide comprising at greater percentage of PD-L1-positive tumor cells than a cutoff value (CoV) is considered QCS-positive. The ODmin and CoV parameters were linked to patient overall survival (OS), by minimizing the Kaplan Meier log-rank p-values and keeping at least 50% prevalence in the QCS-positive subgroup.Fully supervised QCS-IA models were extensively trained using pathologists’ annotations and the performance was validated on unseen data to ensure its generalization and robustness.3 4 The QCS IA was locked and blindly applied on clinical trial data (NCT01693562, durvalumab-treated late-stage NSCLC cohort) without further refinement.ResultsData analytics techniques were used to determine optimal PD-L1 QCS parameters for the clinical trial cohort of N=162 late-stage NSCLC patients. A PD-L1 QCS algorithm (ODmin=8, CoV=57%) is able to stratify durvalumab-treated NSCLC patients at a higher prevalence and more significant log rank p-value (64%, p=0.0001) for OS (figure 1) compared to pathologist TPS (59%, p=0.01). Median OS times of (19.2 months vs 7.9 months) was observed in the QCS-positive vs negative subgroups, respectively. The box plots (figure 2) indicate an overall good agreement (72% concordance) of the fully automated QCS with the pathologists TPS, which quantitatively supports the positive visual assessment of the cell segmentation accuracy.Abstract 365 Figure 1Kaplan Meier (KM) curves for OS stratification. KM curves for Overall Survival (OS) stratification with (left) manual PD-L1 TPS score (25% cutoff), and (right) automated QCS (57% cutoff).Abstract 365 Figure 2QCS scores within TPS positive and negative groups. Box plot indicating percent positive cells (OD≥8) as measured by PD-L1 QCS within the PD-L1 high (red) and low (blue) groups as per pathologist assessment by TPS.ConclusionsThe novel Quantitative Continuous Scoring (QCS) provides an objective way of correlating a quantitative estimate of PD-L1 IHC expression on tumor cells with survival of durvalumab-treated NSCLC patients. This data establishes a first proof-of-concept demonstrating the potential utility of PD-L1 QCS towards precision medicine in immuno-oncology.ReferencesRebelatto M, et al. Development of a programmed cell death ligand-1 immunohistochemical assay validated for analysis of non-small cell lung cancer and head and neck squamous cell carcinoma. Diagnostic Pathology 2016.Tsao M S, et al. PD-L1 immunohistochemistry comparability study in real-life clinical samples: results of blueprint phase 2 project. Journal of Thoracic Oncology 2018.Gustavson M, et al. Novel approach to HER2 quantification: digital pathology coupled with AI-based image and data analysis delivers objective and quantitative HER2 expression analysis for enrichment of responders to trastuzumab deruxtecan (T-DXd; DS-8201), specifically in HER2-low patients. (2021) DOI: 10.1158/1538-7445.SABCS20-PD6-01Kapil A, et al. Domain adaptation-based deep learning for automated tumor cell (TC) scoring and survival analysis on PD-L1 stained tissue images. IEEE Transactions on Medical Imaging DOI: 10.1109/TMI.2021.3081396Ethics ApprovalClinical study NCT01693562, from which data in this report were obtained, was carried out in accordance with the Declaration of Helsinki and Good Clinical Practice guidelines. The study protocol, amendments, and participant informed consent document were approved by the appropriate institutional review boards.


Sign in / Sign up

Export Citation Format

Share Document