ArticlePDF Available

Single-cell RNA sequencing reveals distinct tumor microenvironmental patterns in lung adenocarcinoma

Authors:

Abstract and Figures

Recent developments in immuno-oncology demonstrate that not only cancer cells, but also the tumor microenvironment can guide precision medicine. A comprehensive and in-depth characterization of the tumor microenvironment is challenging since its cell populations are diverse and can be important even if scarce. To identify clinically relevant microenvironmental and cancer features, we applied single-cell RNA sequencing to ten human lung adenocarcinomas and ten normal control tissues. Our analyses revealed heterogeneous carcinoma cell transcriptomes reflecting histological grade and oncogenic pathway activities, and two distinct microenvironmental patterns. The immune-activated CP²E microenvironment was composed of cancer-associated myofibroblasts, proinflammatory monocyte-derived macrophages, plasmacytoid dendritic cells and exhausted CD8+ T cells, and was prognostically unfavorable. In contrast, the inert N³MC microenvironment was characterized by normal-like myofibroblasts, non-inflammatory monocyte-derived macrophages, NK cells, myeloid dendritic cells and conventional T cells, and was associated with a favorable prognosis. Microenvironmental marker genes and signatures identified in single-cell profiles had progonostic value in bulk tumor profiles. In summary, single-cell RNA profiling of lung adenocarcinoma provides additional prognostic information based on the microenvironment, and may help to predict therapy response and to reveal possible target cell populations for future therapeutic approaches.
Composition of the immune microenvironment of lung adenocarcinomas A UMAPs based on the top 20 principal components of all immune single-cell transcriptomes split by tissue type, color-coded by cell cluster; and relative quantification of myeloid and lymphoid cell clusters per tissue type and, for tumor samples, per patient. B Average gene expression of selected marker genes for immune cell clusters, for cell cluster color code see (A). C Module scores of gene signatures related to inflammation and M1/M2 polarization of different macrophage clusters, white and black arrowheads indicate monocyte-derived macrophage clusters 1 and 2, respectively, for cell cluster color code see (A). D Correlation of the relative quantity of selected myeloid immune cell clusters, for patient color code see (G); Spearman’s correlation statistics, linear regression line. E Immunohistochemical staining of CXCL9 and CD123 as markers for monocyte-derived macrophage cluster 2 and plasmacytoid dendritic cells, respectively, quantification of CXCL9+ or CD123+ cells per 0.48 mm², mean ± s.d., n = 10 per patient, for patient color code see (G); Pearson’s correlation statistics and linear regression line using mean values per patient. F Module scores of gene signatures related to cytotoxicity and exhaustion of different CD8+ T cell clusters, white and black arrowheads indicate cell clusters enriched in normal or tumor tissue, respectively, for cell cluster color code see (A). G Correlation of the relative quantity of selected lymphoid and myeloid immune cell clusters, color-coded by patient; Spearman’s correlation statistics, linear regression line.
… 
Tumor microenvironmental patterns in lung adenocarcinomas A Principal component analysis based on the proportion of stromal and immune cell clusters, color-coded by histological subtype, patients indicated. B Normalized proportion of stromal and immune cell clusters, mean module scores of tumor cell signatures, histological subtypes and mutation status per patient, patients sorted along the first principal component from principal component analysis in (A), cell clusters included in the model in (H) in bold. C Correlation of the proportion of stromal and immune cell clusters, most connected section of correlation network plot shown; Spearman’s correlation statistics, only correlations with rho > 0.7 and p < 0.05 shown. A, B, C Cell clusters occurring in <3 patients were excluded from analyses. D Number of potential paracrine interactions from microenvironmental cell clusters to tumor cells of the N³MC or CP²E pattern, computed using CellPhoneDB, grouped by interaction families, color-coded by number of interactions (see also Supplementary Fig. 10). E–G Analysis of the TCGA lung adenocarcinoma cohort. E Correlation of ssGSEA enrichment scores based on marker genes of selected microenvironmental cell clusters and tumor cell signatures; n = 533, Spearman’s correlation statistics, linear regression line. F Kaplan–Meier overall survival curves, cases grouped by the ratio of ssGSEA enrichment scores of indicated microenvironmental cell clusters or tumor cell signatures or a combined signature encompassing all cell clusters of the N³MC or CP²E pattern, respectively; n = 524, log-rank statistics. G Proportion of patients with oncogenic mutations and tumor mutational burden (TMB), patients grouped by ratio of ssGSEA enrichment scores of the combined signature in (F); n = 525 for mutations, Chi-squared test, n = 242 for TMB, two-sided Welch’s t test. H Schematic representation of subtypes of lung adenocarcinoma characterized by different grades of tumor epithelial cell differentiation and different composition of the corresponding tumor microenvironment.
… 
This content is subject to copyright. Terms and conditions apply.
ARTICLE OPEN
Single-cell RNA sequencing reveals distinct tumor
microenvironmental patterns in lung adenocarcinoma
Philip Bischoff
1,2
, Alexandra Trinks
1,2
, Benedikt Obermayer
3
, Jan Patrick Pett
3
, Jennifer Wiederspahn
1,4
, Florian Uhlitz
4,5
,
Xizi Liang
1
, Annika Lehmann
1
, Philipp Jurmeister
1,2,5
, Aron Elsner
6
, Tomasz Dziodzio
2,6
, Jens-Carsten Rückert
6
, Jens Neudecker
6
,
Christine Falk
7,8
, Dieter Beule
3
, Christine Sers
1,2,5
, Markus Morkel
1,2,5
, David Horst
1,2,5
, Nils Blüthgen
1,2,4,5,9
and
Frederick Klauschen
1,5,9
© The Author(s) 2021
Recent developments in immuno-oncology demonstrate that not only cancer cells, but also the tumor microenvironment can guide
precision medicine. A comprehensive and in-depth characterization of the tumor microenvironment is challenging since its cell
populations are diverse and can be important even if scarce. To identify clinically relevant microenvironmental and cancer features, we
applied single-cell RNA sequencing to ten human lung adenocarcinomas and ten normal control tissues. Our analyses revealed
heterogeneous carcinoma cell transcriptomes reecting histological grade and oncogenic pathway activities, and two distinct
microenvironmental patterns. The immune-activated CP²E microenvironment was composed of cancer-associated myobroblasts,
proinammatory monocyte-derived macrophages, plasmacytoid dendritic cells and exhausted CD8+T cells, and was prognostically
unfavorable. In contrast, the inert N³MC microenvironment was characterized by normal-like myobroblasts, non-inammatory
monocyte-derived macrophages, NK cells, myeloid dendritic cells and conventional T cells, and was associated with a favorable prognosis.
Microenvironmental marker genes and signatures identied in single-cell proles had progonostic value in bulk tumor proles. In
summary, single-cell RNA proling of lung adenocarcinoma provides additional prognostic information based on the microenvironment,
and may help to predict therapy response and to reveal possible target cell populations for future therapeutic approaches.
Oncogene (2021) 40:6748–6758; https://doi.org/10.1038/s41388-021-02054-3
INTRODUCTION
Lung cancer has a poor prognosis and accounts for the majority of
new cases and deaths of cancer worldwide [1]. The most common
subtype of lung cancer, in particular in nonsmokers, is lung
adenocarcinoma [2]. Presently, molecular proling of driver
mutations is guiding treatment with targeted therapies [3,4]
and expression of PD-L1 in tumor cells is used to predict response
to immune checkpoint inhibitors [5]. Comprehensive characteriza-
tion including features of the tumor microenvironment could
provide more precise patient stratications.
Cancers are multicellular communities comprising malignant
epithelial cells and different types of nonmalignant immune and
stromal cells which exhibit dynamic and reciprocal interactions.
Modulation of immune responses, remodeling of the extracellular
matrix and neoangionesis essentially determine the aggressive-
ness of cancer [6]. Current bulk omics analyses do not allow high-
resolution characterization of cellular diversity of the tumor
microenvironment. However, comprehensive single-cell proling
of patient tissue is emerging as an essential tool to estimate the
clinical relevance of individual cell types in the tumor.
In this study, we analyzed tumor epithelial cells and associated
nonmalignant cells of the tumor microenvironment of lung
adenocarcinomas by single-cell RNA sequencing. Extending
previous single-cell studies [710], we found that the hetero-
geneous cellular composition of the tumor microenvironment
across patients follows specic patterns that were associated with
the differentiation grade of carcinoma cells. Translation of our
ndings to an retrospective cohort characterized by bulk gene
expression revealed potential prognostic relevance of microenvir-
onmental patterns. We conclude that a comprehensive proling of
the lung adenocarcinoma microenvironment may help to reveal
novel clinically relevant tumor subtypes based on carcinoma cells
and microenvironmental features.
RESULTS
Single-cell RNA sequencing uncovers the cellular diversity of
lung adenocarcinomas
To study the cellular composition of lung adenocarcinoma, ten
normal lung and ten lung adenocarcinoma fresh tissue samples
Received: 26 April 2021 Revised: 16 September 2021 Accepted: 30 September 2021
Published online: 18 October 2021
1
Institute of Pathology, CharitéUniversitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
2
Berlin Institute of
Health, CharitéUniversitätsmedizin Berlin, Berlin, Germany.
3
Core Unit Bioinformatics (CUBI), Berlin Institute of Health at CharitéUniversitätsmedizin Berlin, Berlin, Germany.
4
IRI Life Sciences, Humboldt University of Berlin, Berlin, Germany.
5
German Cancer Consortium (DKTK) Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg,
Germany.
6
Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, CharitéUniversitätsmedizin Berlin, corporate member of Freie Universität Berlin and
Humboldt-Universität zu Berlin, Berlin, Germany.
7
Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany.
8
TTU-IICH Hannover-Braunschweig Site, German
Center for Infectious Diseases (DZIF), Braunschweig, Germany.
9
These authors contributed equally: Nils Blüthgen, Frederick Klauschen. email: philip.bischoff@charite.de
www.nature.com/onc Oncogene
1234567890();,:
Content courtesy of Springer Nature, terms of use apply. Rights reserved
were collected during surgery and subjected to unsorted single-
cell RNA sequencing, yielding 114,489 high-quality transcriptomes
after quality control and ltering (Fig. 1A, Supplementary Fig. 1A,
B). Evaluation of consecutive H&E stained tissue sections showed
tumor morphology ranging from well differentiated lepidic to
poorly differentiated sarcomatoid growth patterns (Supplemen-
tary Fig. 2).
Analysis and visualization by Uniform Manifold Approximation
and Projection (UMAP) [11] showed that single-cell transcriptomes
of different tissue types or patients intermingled in many clusters,
excluding general batch effects, and partly formed tumor- or
patient-specic clusters, indicating underlying biological differ-
ences (Fig. 1B, C, Supplementary Fig. 1C). To uncover which
cellular compartments account most for interpatient variability, we
analyzed single-cell transcriptomes for the expression of epithelial,
immune, and stromal marker genes. In total, 20,450 epithelial,
89,766 immune, and 4273 stromal single-cell transcriptomes were
covered, suggesting an overrepresentation of immune cell
transcriptomes as observed in other studies [9,12]. Yield of
epithelial transcriptomes varied by histological subtype, as we
observed a frequency <10% in solid/sarcomatoid, but up to >40%
in lepidic/acinar carcinomas (Fig. 1D). Epithelial cells showed the
highest degrees of interpatient heterogeneity (Fig. 1C, D).
Intertumoral heterogeneity of tumor epithelial cells reects
differentiation gradients
To further dissect interpatient variability within the epithelial cell
compartment, epithelial transcriptomes were subset and reclus-
tered (Supplementary Fig. 3A). Clusters were dened as normal or
tumor cell clusters, based on tissue origin (Supplementary Fig. 3B),
which was largely congruent with the copy-number status of cells
(Supplementary Fig. 4A, B), and demonstrated a tumor purity
>90%. Within the normal cell clusters, we found alveolar type 1
and 2, club, ciliated, and even a small cluster of neuroendocrine
cells (Fig. 2A), which were characterized by expression of typical
individual marker genes (Fig. 2B) and gene signatures (Supple-
mentary Fig. 5A, B) [13,14]. The club cell cluster also expressed
basal cell marker genes such as NGFR and KRT5 indicating an
admixture of small amounts of basal cells (Fig. 2B). Tumor cell
clusters segregated from normal cell clusters and were mainly
patient-specic, indicating intertumoral heterogeneity (Fig. 2A).
This was underlined by a variety of genes differentially expressed
across tumors such as EGFR, TFF3, CDKN2A, and SFTPA2 (Fig. 2C,
black arrowheads), correlating with protein expression as shown
by immunostaining (Fig. 2D). We quantied oncogenic signal
strengths by pathway target gene signature expression and found
highly variable activities for EGFR, TGFβ, JAK/STAT, Hypoxia, and
PI3K signaling across the different patients (Fig. 2E). These signal
strengths were largely unrelated to the mitotic activity of tumor
epithelial cells (Supplementary Fig. 3C). p53 signaling was
signicantly reduced in tumors harboring TP53 mutations,
whereas pathway activity scores for EGFR and MAPK signaling
were not signicantly higher in KRAS-mutated compared to
KRAS-wildtype tumors (Supplementary Fig. 3D).
Despite obvious intertumoral heterogeneity, we also noted
shared features of epithelial tumor cell transcriptomes across
patient subgroups. In order to emphasize similarities between
tumors, epithelial transcriptomes were embedded in low-
dimensional UMAPs (4, 6, 8 instead of 20 dimensions). Here,
tumor cells clustered by histological subtype rather than by
Fig. 1 Single-cell RNA sequencing of lung adenocarcinomas. A Schematic representation of the workow, ten normal (blue) and ten tumor
(red) tissue samples were obtained from 12 patients. B,CUMAPs based on the top 15 principal components of all single-cell transcriptomes
after ltering, color-coded by (B) tissue type, or (C) patient. DOverview of clinical features, clinically relevant oncogenic mutations and gene
fusions; quantication of main cell types per patient and UMAP of all single-cell transcriptomes color-coded by main cell type.
P. Bischoff et al.
6749
Oncogene (2021) 40:6748 – 6758
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Fig. 2 Intertumoral heterogeneity of tumor epithelial cells in lung adenocarcinomas. A UMAPs based on the top 20 principal components
of all epithelial single-cell transcriptomes color-coded by tissue type, cell type and patient, and quantication of epithelial cell types per tissue
type, AT1, alveolar type 1 cells, AT2, alveolar type 2 cells. BAverage gene expression of selected marker genes for normal epithelial cell types.
CDifferentially expressed genes in tumor epithelial cells grouped by patients, maximum top ten genes showed per patient, for patient color
code see (A). DImmunohistochemical staining of proteins encoded by selected differentially expressed genes indicated by black arrowheads
in (C). EMean pathway activity scores of tumor epithelial cells grouped by patient. FDistribution of histological subtypes, (G) mean module
scores of normal epithelial cell type gene signatures, and (H) mean pathway activity scores of tumor epithelial cells sorted along principal
component 1 (PC1). F,G,HPrincipal component analysis based on gene expression of all tumor epithelial single-cell transcriptomes;
schematic depiction of tumor cell signature module scores along PC1.
P. Bischoff et al.
6750
Oncogene (2021) 40:6748 – 6758
Content courtesy of Springer Nature, terms of use apply. Rights reserved
patient (Supplementary Fig. 3E), and the rst principal component
(PC1) displayed a gradient along histological grades (Fig. 2F,
Supplementary Fig. 3E, F). Interestingly, SCGB3A1 and SCGB3A2
(Fig. 2C, white arrowheads), two genes that were previously
associated with lung development [15], were positively correlated
with PC1 (Supplementary Fig. 3G, arrowheads). Moreover, gene
signature scores of normal lung cell types [13] along PC1 showed
a strong positive correlation with gene expression proles of
alveolar type 1 and 2 as well as club cells (Fig. 2G). Together, this
indicates that PC1 reects the degree of differentiation of tumor
epithelial cells. Hence, the top 30 genes positively and negatively
correlated with PC1 were dened as an alveolar/club-likeand
undifferentiatedtumor cell signature, respectively (Fig. 2FH,
Supplementary Fig. 3G, H). While tumor cells with different
degrees of differentiation exhibited no clear differences in mitotic
activity (Supplementary Fig. 3I), we found high pathway activity
scores for JAK/STAT, Hypoxia, EGFR and TGFβsignaling in
undifferentiated, and high scores for PI3K signaling in alveo-
lar/club cell-liketumor epithelial cells, respectively (Fig. 2H). We
conclude that tumor epithelial cells of different lung adenocarci-
noma patients exhibit transcriptional patterns along a spectrum
ranging from undifferentiated to alveolar/club cell-like pheno-
types correlating with distinct oncogenic pathway activity.
Two subtypes of myobroblasts constitute the tumor stromal
microenvironment
We subset and analyzed stromal cells from both normal
and tumor tissue samples. Different clusters of endothelial and
lymphatic endothelial cells, broblasts, myobroblasts and
smooth muscle cells and mesothelial cells (Fig. 3A, Supplementary
Fig. 6A) were identied by marker genes (Fig. 3B) and gene
signatures (Supplementary Fig. 5A, B) [13,14]. Tumor endothelial
cells were mainly represented by clusters 2 and 4 (Fig. 3A), and
showed high expression of angiogenesis markers such as VWA1
and HSPG2, as well as INSR, encoding an endothelial marker
protein and possible therapeutic target [9] (Supplementary Fig. 6B,
arrowheads).
Among the broblastic/muscle cell clusters, we detected a shift
from broblast to myobroblast cell clusters in tumor tissues
(Fig. 3A), which we also observed in an independent dataset
(Supplementary Fig. 7A). Myobroblast clusters were characterized
by expression of both broblastic marker genes, such as PDGFRA
and LUM, and smooth muscle marker genes, such as MYLK and
ACTA2 (Fig. 3B). Notably, myobroblast cluster 2 was almost
exclusively found in tumor tissues while myobroblast cluster 1
encompassed normal and tumor tissues. Myobroblast cluster 2
displayed high expression of collagens such as COL3A1,COL5A1,
Fig. 3 Composition of the stromal microenvironment of lung adenocarcinomas. A UMAPs based on the top 20 principal components of
all stromal single-cell transcriptomes split by tissue type, color-coded by cell cluster; and relative quantication of endothelial and
broblastic/muscle cell clusters per tissue type and, for tumor samples, per patient. BAverage gene expression of selected marker genes
for stromal cell clusters, for cell cluster color code see (A). CDifferentially expressed genes of broblastic/muscle cell clusters, maximum top
ten genes showed per cell cluster, for cell cluster color code see (A), black arrowheads indicate relevant marker genes of myobroblast
cluster 2 mentioned in the main text. DMean pathway activity scores of different broblastic/muscle cell clusters, mesothelial cells
excluded, black arrowheads indicate relevant pathways of myobroblast clusters 1 and 2 mentioned in the main text. ECorrelation of the
relative quantity of myobroblast clusters 1 and 2, color-coded by patient; Spearmans correlation statistics, linear regression line.
FImmunohistochemical staining of CTHRC1 as marker for myobroblast cluster 2 (see also (C)), quantication of proportion of stromal
areal covered by CTHRC1+cells, mean ± s.d., n=10 per patient, for patient color code see (E); Pearsons correlation statistics and linear
regression line using mean values per patient.
P. Bischoff et al.
6751
Oncogene (2021) 40:6748 – 6758
Content courtesy of Springer Nature, terms of use apply. Rights reserved
COL5A2 and COL6A3, other matrix proteins such as VCAN,aswellas
matrix-degrading enzymes such as SULF1 and MMP11, suggesting
roles in extracellular matrix remodeling (Fig. 3C, arrowheads).
Myobroblast cluster 2 was also characterized by high activity of
TGFβand JAK/STAT signaling as well as hypoxia-induced pathways
(Fig. 3D), which are known features of cancer-associated myobro-
blasts [16,17]. In contrast, myobroblast cluster 1 exhibited low
activities of these pathways. Relative proportions of myobroblast
clusters 1 and 2 within the broblastic/muscle cell compartment
correlated inversely across patients (Fig. 3E). The distribution of
myobroblast cluster 2 cells could be validated by immunostaining
for the cluster-specicmarkerCTHRC1(Fig.3F, see also Fig. 3C). We
conclude that myobroblasts cluster 1 and 2 represent normal-like
and cancer-associatedphenotypes of myobroblasts, respectively,
and either of them can predominate the stromal microenvironment.
The tumor immune microenvironment exhibits pro- and non-
inammatory traits
We next subset and analyzed immune cells of the tumor
microenvironment. We identied different clusters of tissue-
resident and monocyte-derived macrophages, monocytes, mye-
loid and plasmacytoid dendritic cells, mast cells, and T, NK, B,
and plasma cells (Fig. 4A, Supplementary Fig. 8AC) based on
typical marker genes (Fig. 4B) and gene signatures (Supplemen-
tary Fig. 5A, B) [13,14].
Although immune cell clusters usually encompassed both normal
and tumor tissue-derived transcriptomes, we noted quantitative
shifts in the cellular composition of the tumor immune microenvir-
onment (Fig. 4A). Among myeloid cell types in the tumor,
monocyte-derived macrophages, dened by markers such as
CD14, CSF1R and LGMN (Fig. 4B) [18,19] and dendritic cells were
increased, while tissue-resident macrophages and monocytes were
decreased, which we also observed in an independent dataset
(Supplementary Fig. 7B). Tumor myeloid cell compartments were
particularly rich in monocyte-derived macrophage clusters 1 and 2.
Of these, cluster 1 showed high expression of SELENOP (Supple-
mentary Fig. 8B, arrowhead), which has been related to M2
polarization [20], and low scores of immune response-related
signatures (Fig. 4C, white arrowheads), while cluster 2 expressed
high levels of proinammatory chemokines, such as CXCL9 and
CXCL10, the proinammatory cytokine IL1B (Supplementary Fig. 8B,
arrowheads), and gene signatures related to immune response and
M1 polarization [21,22](Fig.4C, black arrowheads). This indicates
that monocyte-derived macrophage clusters 1 and 2 represent non-
and proinammatory functional states, respectively, which were
inversely correlated across patients (Fig. 4D). In addition, the
proportion of proinammatory monocyte-derived macrophages
cluster 2 correlated with other myeloid cell types such as
plasmacytoid dendritic cells (Fig. 4D). The distribution of proin-
ammatory monocyte-derived macrophages cluster 2 and plasma-
cytoid dendritic cells was validated by immunostaining for CXCL9
and CD123, respectively (Fig. 4E, Supplementary Fig. 8D).
Within the tumor lymphoid cell compartment, CD8+T, B, and
plasma cells were increased, while NK and conventional T cells
were decreased compared to normal tissue controls (Fig. 4A).
Regulatory T cells were almost exclusively found in tumor tissue
samples and expressed inhibitory molecules such as CTLA4 and
TIGIT (Supplementary Fig. 8C, arrowheads) corresponding to their
immunosuppressive role [23]. We identied in total four clusters of
CD8+T cells (Fig. 4A). The tumor-enriched CD8+T cell clusters 1
and 2 and proliferating CD8+T cells were characterized by an
exhaustion signature [10] (Fig. 4F, black arrowheads), while this
score was low in the normal-enriched CD8+T cell cluster 3
(Fig. 4F, white arrowhead). Exhausted CD8+T cells were
signicantly associated with proinammatory monocyte-derived
macrophages (cluster 2) and plasmacytoid dendritic cell numbers
(Fig. 4G), and a similar trend, albeit not signicant, was seen for
regulatory T cells.
Taken together, we identify patient-overarching changes of the
immune cell composition from normal lung tissue to adenocarci-
noma, and distinct tumor immune microenvironment patterns
contributing to interpatient heterogeneity.
The tumor microenvironment of lung adenocarcinoma
features two major patterns
To integrate our analyses of variable cell prevalences in the tumor
microenvironment, we calculated proportions of cells of the
myeloid, lymphoid, endothelial and broblastic/muscle cell
compartments across patients (for cell counts see Supplementary
Tables 14). Principal component analysis showed that tumors
formed subgroups based on the cellular composition of the tumor
microenvironment (Fig. 5A). One group of tumors (P018, P019,
P024, P031, P032, and P033) was marked by normal-like
myobroblasts, non-inammatory monocyte-derived macro-
phages, NK cells, myeloid dendritic cells and conventional
T cells, referred to as N³MC pattern (Fig. 5B, Supplementary Figs.
7C, 9A). A second group of tumors (P023, P027, P030 and P034)
was characterized by cancer-associated myobroblasts, proin-
ammatory monocyte-derived macrophages, plasmacytoid den-
dritic cells and exhausted CD8+T cells, referred to as CP²E pattern
(Fig. 5B, Supplementary Figs. 7C, 9A). Comparable patterns were
also found in an independent single-cell dataset of primary lung
adenocarcinoma (Supplementary Fig. 7DF). The two microenvir-
onmental patterns roughly separated tumors by histological grade
(Fig. 5A, Supplementary Fig. 7C), by expression scores of alveolar/
club-likeversus undifferentiatedtumor cell signatures (Fig. 5B),
and by prevalence of microenvironmental cell clusters (Fig. 5B, C,
Supplementary Fig. 9B).
We quantied potential receptor-ligand interactions within the
N³MC and CP²E tumor microenvironments, which were most
frequent between tumor cells and cancer-associated myobro-
blast cluster 2 of the CP²E pattern (Supplementary Table 5).
Focusing on the most relevant oncogenic pathways (Supplemen-
tary Table 6), we found that tumor cells in the CP²E environment
receive potential paracrine signals from cancer-associated myo-
broblast cluster 2 activating Ephrin, FGF, WNT, TGFβ, and BMP
signaling, and from proinammatory monocyte-derived macro-
phages cluster 2 potentially activating JAK/STAT signaling (Fig. 5D,
Supplementary Fig. 10).
To test if tumor microenvironmental patterns can be recapitu-
lated by bulk gene expression patterns in larger patient cohorts,
we analyzed the expression of cell cluster marker genes in The
Cancer Genome Atlas (TCGA) lung adenocarcinoma cohort
encompassing 533 patients (for marker genes see Supplementary
Tables 710). We found a specic positive correlation of the
alveolar/club-like tumor cell signature with non-inammatory
monocyte-derived macrophages and normal-like myobroblasts,
recapitulating the N³MC pattern, and of the undifferentiated
tumor cell signature with proinammatory monocyte-derived
macrophages and cancer-associated myobroblasts, recapitulat-
ing the CP²E pattern (Fig. 5E, Supplementary Fig. 9C).
To investigate the biological and clinical relevance of these
patient subgroups, we analyzed the overall survival of the TCGA
lung adenocarcinoma cohort contigent on expression of the
different gene signatures. N³MC-related gene signatures were
associated with a better overall survival compared to signatures of
the CP²E pattern (Fig. 5F). In multivariate analyses, this association
was signicant for the tumor cell signatures (HR 0.50, 95% CI 0.35-
0.71, p=0.0001). Although individual cell cluster signatures had
different prognostic relevance, a combination of all cell cluster
signatures as well as a simplied signature of 20 genes separated
patients into prognostic subgroups (Fig. 5F, Supplementary Fig.
11AC, Supplementary Table 11). The group with worse prognosis
and higher CP²E scores was characterized by more frequent
mutations in KEAP,NTRK3,ROS1, and TP53 as well as higher
mutational burden (Fig. 5G).
P. Bischoff et al.
6752
Oncogene (2021) 40:6748 – 6758
Content courtesy of Springer Nature, terms of use apply. Rights reserved
In summary, our results show that lung adenocarcinomas can
be stratied into clinically relevant subgroups which are not only
characterized by the grade of tumor epithelial cells, but also by
the cellular composition of their associated tumor microenviron-
ment (Fig. 5H). We therefore propose two major microenviron-
mental patterns in lung adenocarcinoma which we term N³MC
and CP²E as akronyms of the respective characteristic cell types.
DISCUSSION
By applying single-cell RNA sequencing to lung adenocarcinomas,
we identied two major microenvironmental patterns, referred to
as N³MC and CP²E (Fig. 5H). We characterized the N³MC tumor
microenvironment as rather inert and normal-like, as it was
dened by normal-like myobroblasts, conventional T cells, NK
cells, non-inammatory monocyte-derived macrophages and
P. Bischoff et al.
6753
Oncogene (2021) 40:6748 – 6758
Content courtesy of Springer Nature, terms of use apply. Rights reserved
myeloid dendritic cells. The N³MC pattern was associated with an
alveolar/club-like gene expression pattern of carcinoma cells,
lower histological grade and better prognosis. In contrast, the CP²E
tumor microenvironment was proinammatory and characterized
by proinammatory monocyte-derived macrophages, plasmacy-
toid dendritic cells, exhausted CD8+T cells and cancer-associated
myobroblasts. The CP²E pattern was associated with an
undifferentiated gene expression pattern of carcinoma cells,
higher histological grade, and worse prognosis.
In the clinic, tumor stage and histological grading are used to
predict prognosis in lung adenocarcinoma. Grading is based on
the predominant growth pattern but other histological features
have been proposed in order to improve risk stratication [24,25].
However, these concepts focus on the carcinoma cells and do not
include features of the complex composition of the tumor
microenvironment. Most available studies on prognostic micro-
environmental features focus on single-cell types identied by
immunostaining. Cancer-associated broblasts were found to
correlate with worse prognosis using different biomarkers
[2628], while tumor-inltrating T cells indicated better prognosis
in multiple studies [2931]. The prognostic value of proinamma-
tory tumor-associated macrophages (M1) is inconclusive across
the available studies, likely because differentiation of macrophage
subtypes by immunostaining is limited [32]. In order to account for
the complexity of the tumor microenvironment, patterns based on
tumor-inltrating T cells and PD-L1 expression have been
proposed [33,34]. Beyond this, the cellular complexity has
recently been studied by multiplexed immunostaining [35,36].
Gene expression proling of immune-related genes provide
further functional insights, but have yet been limited to the use
of bulk methods [3739]. Single-cell RNA sequencing combines
both a comprehensive census of microenvironmental cell types
and an in-depth functional characterization of their transcriptional
proles. In our study, the microenvironmental CP²E pattern was
associated with worse prognosis, which is in line with existing
evidence for cancer-associated myobroblasts [40] and plasma-
cytoid dendritic cells [41], but partly contradicts previous works
with regard to proinammatory tumor-associated macrophages
[32]. The absence of cancer-associated myobroblasts, and the
presence of NK cells [42] and conventional T cells [31] support the
association of the N³MC pattern with better prognosis.
Characterization of PD-L1 expression on tumor-inltrating
immune cells is already used to predict response to immune
checkpoint inhibitors [43], but more comprehensive character-
ization of the microenvironment will likely provide even deeper
insights for patient stratication and drug development.
Recently, expression of the proinammatory cytokines CXCL9
and CXCL10 by tumor-associated macrophages has been shown
to be essential for tumor response to anti-PD-L1 therapy [44,45].
High expression of these cytokines in proinammatory
monocyte-derived macrophages, high numbers of exhausted
CD8+T cells [46] and high mutational burden [47] indicates a
predictive relevance of the CP²E microenvironment. In addition,
the CP²E pattern was characterized by cancer-associated
myobroblasts expressing potential novel therapeutic targets,
such as PSTN and MMP11 [48,49]. Despite an overall depletion of
NK cells in tumors in agreement with other studies [7,9], the
N³MC microenvironmental pattern still contained small popula-
tions of NK cells which could potentially be targeted by
immunostimulatory agents [50]. Furthermore, we conrmed
enrichment of B cells in some tumors [8,9]withpotential
importance in development of novel immunotherapies [51].
We envisage that single-cell approaches will also contribute to a
better understanding of tumor cells with regard to their high
mutational burden [52] and genomic intertumoral heterogeneity
[53]. Squamous and adenocarcinomas of the lung can exhibit
distinct transcriptional differences [54,55], and interestingly,
despite intertumoral heterogeneity, we found that tumor epithe-
lial transcriptomes retained histology-related patterns that we
termed alveolar/club-likeand undifferentiatedacross patients
which are reminiscent of recently dened signatures [9]. The
undifferentiatedtumor cell signature was characterized by high
activity of TGFβsignaling and hypoxia-induced pathways, which
are both known activators of epithelial-mesenchymal transition
[56,57], corresponding to poor histological differentiation of these
tumors. The alveolar/club-likesignature was characterized by
PI3K signaling, an important oncogenic pathway promoting tumor
growth and potentially affecting the tumor microenvironment by
upregulating PD-L1 expression in carcinoma cells [5860]. EGFR
and JAK/STAT signaling, associated with the undifferentiated
signature, and PI3K signaling, associated with the alveolar/club-
likesignature, are potential therapeutic targets [4,60,61]. We
identied cancer-associated myobroblasts as potential sources of
TGFβligands in the CP²E pattern, while EGFR interactions were
equally present in both patterns, underlining that signaling
pathways in tumor cells can be activated extrinsically via paracrine
signals from the microenvironment but also via other mechan-
isms, e.g., intrinsically via oncogenic mutations.
Single-cell approaches provide high-resolution information on
the cellular complexity of tumor tissues but are limited in cohort
size. We validated some ndings from single-cell RNA sequencing
by immunostainings and identied a prognostic set of 20 genes
recapitulating microenvironmental patterns. Alternative methods,
such as multiplex immunostaining or bulk gene expression
proling, will facilitate validation of ndings in large cohorts and
translation into clinical application. So far, our ndings suggest a
prognostic relevance of tumor microenvironmental patterns
which could aid in therapeutical decision making. Moreover,
preclinical ndings from other groups suggest that tumor
microenvironmental patterns could be associated with differential
responses to immune checkpoint inhibition, thus distinct micro-
environmental features such as tumor-inltrating CD8+T cells or
CXCL9+macrophages could complement current patient strati-
cation by PD-L1 expression. Finally, characterization of the tumor
microenvironment on single-cell resolution can provide insight on
possible novel therapeutic targets. It remains an open question to
which extent cancer cells shape their microenvironment and to
which extent the microenvironment affects cancer cells. Preclinical
efforts need to be complemented by translational studies to
identify critical mechanisms in this complex network that
Fig. 4 Composition of the immune microenvironment of lung adenocarcinomas. A UMAPs based on the top 20 principal components of all
immune single-cell transcriptomes split by tissue type, color-coded by cell cluster; and relative quantication of myeloid and lymphoid cell
clusters per tissue type and, for tumor samples, per patient. BAverage gene expression of selected marker genes for immune cell clusters, for
cell cluster color code see (A). CModule scores of gene signatures related to inammation and M1/M2 polarization of different macrophage
clusters, white and black arrowheads indicate monocyte-derived macrophage clusters 1 and 2, respectively, for cell cluster color code see
(A). DCorrelation of the relative quantity of selected myeloid immune cell clusters, for patient color code see (G); Spearmans correlation
statistics, linear regression line. EImmunohistochemical staining of CXCL9 and CD123 as markers for monocyte-derived macrophage cluster 2
and plasmacytoid dendritic cells, respectively, quantication of CXCL9+or CD123+cells per 0.48 mm², mean ± s.d., n=10 per patient, for
patient color code see (G); Pearsons correlation statistics and linear regression line using mean values per patient. FModule scores of gene
signatures related to cytotoxicity and exhaustion of different CD8+T cell clusters, white and black arrowheads indicate cell clusters enriched
in normal or tumor tissue, respectively, for cell cluster color code see (A). GCorrelation of the relative quantity of selected lymphoid and
myeloid immune cell clusters, color-coded by patient; Spearmans correlation statistics, linear regression line.
P. Bischoff et al.
6754
Oncogene (2021) 40:6748 – 6758
Content courtesy of Springer Nature, terms of use apply. Rights reserved
determine tumor response to targeted or immune therapies in the
clinical context. While our study demonstrates how single-cell
gene expression proling of clinical samples can contribute to this
task, in the future, other single-cell approaches comprising spatial
information [62], surface protein expression [63], and epigenetic
characterization [64] will help to complete the picture [65].
METHODS
Collection of tissue specimens
Fresh normal lung parenchyma and tumor tissues were obtained during
intraoperative pathologist consultation from previously untreated lung
adenocarcinoma patients undergoing primary surgery. Patients were
aware of the planned research and agreed to the use of tissue. Research
was approved by vote EA4/164/19 of the ethics committee of Charité
Universitätsmedizin Berlin.
Tissue dissociation and single-cell isolation
Tissue specimens of ~0.10.5 cm³ were stored on ice in Tissue Storage
Solution (Miltenyi, Bergisch Gladbach, Germany) for transport. Tissues were
minced, dissociated using the Tumor Dissociation Kit, human (Miltenyi) and a
gentleMACS Octo Dissociator with heaters (Miltenyi), using program
37C_h_TDK_1 for 3045 min. Subsequent steps were performed at 4 °C. Cell
suspensions were ltered using 100 μm lters, pelleted by centrifugation at
300 × gin BSA-coated low-binding tubes, treated with 1 ml ACK erythrocyte
lysis buffer for 1 min, washed with DMEM, pelleted, resuspended in PBS,
P. Bischoff et al.
6755
Oncogene (2021) 40:6748 – 6758
Content courtesy of Springer Nature, terms of use apply. Rights reserved
ltered using 20 μm lters, debris was removed using the Debris Removal
Solution (Miltenyi), and cells were counted using a Neubauer chamber.
Single-cell RNA sequencing
10,000 single cells were used for single-cell library production, using the
Chromium Single Cell 3Reagent Kit v3 and the Chromium Controller (10x
Genomics, Pleasanton, California, USA) according to the manufacturers
protocol. Libraries were sequenced on a HiSeq 4000 Sequencer (Illumina,
San Diego, California, USA) at in average 54,000 reads per cell.
H&E and immunostaining
35 μm tissue sections of formalin-xed and parafn-embedded (FFPE) tissue
were prepared. For H&E staining, sections were stained for 8 min in acidic
haemalum staining solution (Waldeck, Münster, Germany) and for 2.5 min in
eosin staining solution (Sigma-Aldrich, St. Louis, Missouri, USA) using a Tissue-
Tek Prisma Plus slide stainer (Sakura, Staufen im Breisgau, Germany).
Immunohistochemical stainings were performed on the BenchMark XT
immunostainer (Ventana, Oro Valley, Arizona, USA). Tissue sections were
incubated in CC1 or CC2 buffer (Ventana) for 30 min at 100 °C, and
subsequently with primary antibodies for 60 min and with secondary
antibodies for 30 min at room temperature. Multiplex immunouorescence
stainings were performed using t-CyCIF as described in [66]. Primary
antibodies are listed in Supplementary Table 12. H&E and immunohisto-
chemical images were taken using a Pannoramic SCAN 150 scanner
(3DHISTECH, Budapest, Hungary), immunouorescence images were taken
using a CQ1 microscope (Yokogawa, Musashino, Japan). For quantication,
10 random areas of 0.48 mm² each were analyzed by a pathologist. CD123-
and CXCL9-positve cells were counted per area. For CTHRC1 quantication,
using the ImageJ software, stromal area was manually marked as region of
interest, color channels were split by Color deconvolution, the DAB
channel was binarized using an intensity threshold of 150, and the
proportion of area covered by positive signal was measured per image.
Panel sequencing
For panel sequencing, tumor-enriched areas were macrodissected from
FFPE tissue sections.
DNA was isolated using the Maxwell RSC DNA FFPE Kit (Promega,
Madison, Wisconsin, USA) on a Maxwell RSC 48 Instrument (Promega) and
analyzed using the nNGM panel v1 (ThermoFisher, Waltham, Massachu-
setts, USA), an Ion 530 chip (ThermoFisher) and the Ion Chef/Ion S5 XL
System (ThermoFisher).
RNA was isolated using the Maxwell RSC RNA FFPE Kit (Promega) on a
Maxwell RSC 48 Instrument (Promega) and analyzed using the Oncomine
Focus RNA Assay (ThermoFisher), an Ion 530 chip and the Ion Chef/Ion S5
XL System (ThermoFisher).
The Sequence Pilot Software (Version 4.4.0, JSI Medical Systems) and the Ion
Reporter Software (Version 5.12, ThermoFisher) were used for variant calling.
Single-cell RNA sequencing data analysis
Preprocessing, ltering, and normalization. UMIs were quantied using
Cellranger 3.0.2 (10x Genomics) with reference transcriptome GRCh38.
Subsequent analyses were performed using Seurat v3[67], if not stated
otherwise. Single-cell gene expression data of all patients were merged,
and transcriptomes were ltered for cells with 50010,000 genes detected,
1000100,000 UMIs counted, fraction of mitochondrial reads <30%, and
fraction of hemoglobin reads <5%. After ltering, UMI counts were
variance-stabilized using scTransform with 3000 variable features [68],
while regressing out number of UMIs and fraction of mitochondrial reads.
Clustering and cell type annotation. Top 15 principal components were
used to construct SNN graph and UMAP embedding. Main cell types were
identied by scoring canonical cell type markers across clusters. PCA, SNN
graph construction and UMAP embedding was rerun on main cell type
subsets. Cell type markers were adapted from Habermann et al. [69]. and
Tata and Rajagopal [70] (Supplementary Table 13). Cell type signatures
from Vieira Braga et al. [13] and Travaglini et al. [14] were used to validate
manual cell type annotation. Epithelial or immune contaminated clusters
were identied by EPCAM or PTPRC, respectively, and removed before
further analyses. DoubletFinder v2.0 was used to estimate cell doublet
distribution (Supplementary Fig. 1D). Epithelial cell clusters overrepre-
sented in tumor tissue samples were annotated as tumor cells. For copy-
number assignment, InferCNV v1.3.3 was used with default parameters.
Differential gene expression analysis. Marker genes for each cell cluster
versus all cells of the respective subset were computed using the
FindAllMarkers function with the following parameters: only positive
markers, fraction of expressing cells inside the cluster 0.25, difference
between fraction of expressing cells inside and outside the cluster 0.25,
log fold change between cells inside and outside the cluster 0.25.
Functional analysis. Cell cycle phases were scored as implemented in
Seurat v3. Expression of gene sets of the Hallmark signature collection of
the Broad Institute [21], and M1 vs. M2 up- and downregulated genes [22]
were scored using the AddModuleScore function. Oncogenic signaling
pathway activity scores were computed using the R package progeny
[71,72]. Potential paracrine interactions were computed using the
CellPhoneDBtoolkit with default parameters [73].
Analysis of the TCGA lung adenocarcinoma cohort
FPKM-normalized gene expression values of 533 lung adenocarcinoma
(LUAD) cases were downloaded using the R package TCGAbiolinks[74]and
log2 transformed. Marker genes from myeloid, lymphoid, endothelial, stromal
cell subsets, and the alveolar/club-likeand undifferentiatedtumor cell
signatures were used as gene sets to perform single-sample gene set
enrichment analysis (ssGSEA) [75]onTCGALUADgeneexpressiondatausing
the R package GSVA[76]. Associations between enrichment scores (ES)
from ssGSEA were calculated using the R package corrplot.
Next, patients were grouped by dichotomization of ssGSEA ES (> or
median). Survival data of the TCGA LUAD cohort was available for 524
cases and downloaded using the R package TCGAbiolinks[74]. Survival
curves, log-rank statistics and Cox regression were calculated using the R
packages survivaland survminer. Data on mutations and mutational
burden was available for 525 and 242 cases, respectively.
Statistical analysis
Patient groups were compared using the Welchsttest or MannWhitney
Utest (both two-sided, unequal variances), as indicated, after testing for
normal distribution using the ShapiroWilks test. Distribution of mutations
across patient groups was analyzed using the Chi-squared test. For
correlation analysis, we calculated the Pearson or Spearman correlation
coefcient, as indicated. Pvalues < 0.05 were considered signicant.
Fig. 5 Tumor microenvironmental patterns in lung adenocarcinomas. A Principal component analysis based on the proportion of stromal
and immune cell clusters, color-coded by histological subtype, patients indicated. BNormalized proportion of stromal and immune cell
clusters, mean module scores of tumor cell signatures, histological subtypes and mutation status per patient, patients sorted along the rst
principal component from principal component analysis in (A), cell clusters included in the model in (H) in bold. CCorrelation of the
proportion of stromal and immune cell clusters, most connected section of correlation network plot shown; Spearmans correlation statistics,
only correlations with rho > 0.7 and p< 0.05 shown. A,B,CCell clusters occurring in <3 patients were excluded from analyses. DNumber of
potential paracrine interactions from microenvironmental cell clusters to tumor cells of the N³MC or CP²E pattern, computed using
CellPhoneDB, grouped by interaction families, color-coded by number of interactions (see also Supplementary Fig. 10). EGAnalysis of the
TCGA lung adenocarcinoma cohort. ECorrelation of ssGSEA enrichment scores based on marker genes of selected microenvironmental cell
clusters and tumor cell signatures; n=533, Spearmans correlation statistics, linear regression line. FKaplanMeier overall survival curves,
cases grouped by the ratio of ssGSEA enrichment scores of indicated microenvironmental cell clusters or tumor cell signatures or a combined
signature encompassing all cell clusters of the N³MC or CP²E pattern, respectively; n=524, log-rank statistics. GProportion of patients with
oncogenic mutations and tumor mutational burden (TMB), patients grouped by ratio of ssGSEA enrichment scores of the combined signature
in (F); n=525 for mutations, Chi-squared test, n=242 for TMB, two-sided Welchsttest. HSchematic representation of subtypes of lung
adenocarcinoma characterized by different grades of tumor epithelial cell differentiation and different composition of the corresponding
tumor microenvironment.
P. Bischoff et al.
6756
Oncogene (2021) 40:6748 – 6758
Content courtesy of Springer Nature, terms of use apply. Rights reserved
DATA AVAILABILITY
Processed count data used for analyses is available as a Code Ocean capsule from
https://doi.org/10.24433/CO.0121060.v1.
CODE AVAILABILITY
Code used for analyses is available as a Code Ocean capsule from https://doi.org/
10.24433/CO.0121060.v1. Complementary code is available from https://github.com/
bischofp/single_cell_lung_adenocarcinoma.
REFERENCES
1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer
statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36
cancers in 185 countries. CA Cancer J Clin. 2018;68:394424.
2. Wakelee HA, Chang ET, Gomez SL, Keegan TH, Feskanich D, Clarke CA, et al. Lung
cancer incidence in never smokers. J Clin Oncol. 2007;25:4728.
3. Cancer Genome Atlas Research N. Comprehensive molecular proling of lung
adenocarcinoma. Nature. 2014;511:54350.
4. Hirsch FR, ScagliottiGV, Mulshine JL, Kwon R, Curran WJ Jr, Wu YL, et al. Lung cancer:
current therapies and new targeted treatments. Lancet. 2017;389:299311.
5. Memon H, Patel BM. Immune checkpoint inhibitors in non-small cell lung cancer:
a birds eye view. Life Sci. 2019;233:116713.
6. Altorki NK, Markowitz GJ, Gao D, Port JL, Saxena A, Stiles B, et al. The lung
microenvironment: an important regulator of tumour growth and metastasis. Nat
Rev Cancer. 2019;19:931.
7. Lavin Y, Kobayashi S, Leader A, Amir ED, Elefant N, Bigenwald C, et al. Innate
immune landscape in early lung adenocarcinoma by paired single-cell analyses.
Cell. 2017;169:75065.e17.
8. Lambrechts D, Wauters E, Boeckx B, Aibar S, Nittner D, Burton O, et al. Phenotype
molding of stromal cells in the lung tumor microenvironment. Nat Med.
2018;24:127789.
9. Kim N, Kim HK, Lee K, Hong Y, Cho JH, Choi JW, et al. Single-cell RNA sequencing
demonstrates the molecular and cellular reprogramming of metastatic lung
adenocarcinoma. Nat Commun. 2020;11:2285.
10. Guo X, Zhang Y, Zheng L, Zheng C, Song J, Zhang Q, et al. Global characterization
of T cells in non-small-cell lung cancer by single-cell sequencing. Nat Med.
2018;24:97885.
11. Becht E, McInnes L, Healy J, Dutertre CA, Kwok IWH, Ng LG, et al. Dimensionality
reduction for visualizing single-cell data using UMAP. Nat Biotechnol. 2019;37:3844.
12. Lee HO, Hong Y, Etlioglu HE, Cho YB, Pomella V, Van den Bosch B, et al. Lineage-
dependent gene expression programs inuence the immune landscape of col-
orectal cancer. Nat Genet. 2020;52:594603.
13. Vieira Braga FA, Kar G, Berg M, Carpaij OA, Polanski K, Simon LM, et al. A cellular
census of human lungs identies novel cell states in health and in asthma. Nat
Med. 2019;25:115363.
14. Travaglini KJ, Nabhan AN, Penland L, Sinha R, Gillich A, Sit RV, et al. A molecular cell
atlas of the human lung from single-cell RNA sequencing. Nature. 2020;587:619625.
15. Naizhen X, Kido T, Yokoyama S, Linnoila RI, Kimura S. Spatiotemporal expression
of three secretoglobin proteins, SCGB1A1, SCGB3A1, and SCGB3A2, in mouse
airway epithelia. J Histochem Cytochem. 2019;67:45363.
16. Yoshida GJ. Regulation of heterogeneous cancer-associated broblasts: the mole-
cular pathology of activated signaling pathways. J Exp Clin Cancer Res. 2020;39:112.
17. Petrova V, Annicchiarico-Petruzzelli M, Melino G, Amelio I. The hypoxic tumour
microenvironment. Oncogenesis. 2018;7:10.
18. Franklin RA, Li MO. Ontogeny of tumor-associated macrophages and its impli-
cation in cancer regulation. Trends Cancer 2016;2:2034.
19. Solberg R, Smith R, Almlof M, Tewolde E, Nilsen H, Johansen HT. Legumain
expression, activity and secretion are increased during monocyte-to-macrophage
differentiation and inhibited by atorvastatin. Biol Chem. 2015;396:7180.
20. Solinas G, Schiarea S, Liguori M, Fabbri M, Pesce S, Zammataro L, et al. Tumor-
conditioned macrophages secrete migration-stimulating factor: a new marker for
M2-polarization, inuencing tumor cell motility. J Immunol. 2010;185:64252.
21. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The
Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst.
2015;1:41725.
22. Martinez FO, Gordon S, Locati M, Mantovani A. Transcriptional proling of the
human monocyte-to-macrophage differentiation and polarization: new mole-
cules and patterns of gene expression. J Immunol. 2006;177:730311.
23. Jiang Y, Li Y, Zhu B. T-cell exhaustion in the tumor microenvironment. Cell Death
Dis. 2015;6:e1792.
24. Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, et al. The
2015 World Health Organization classication of lung tumors: impact of genetic,
clinical and radiologic advances since the 2004 classication. J Thorac Oncol.
2015;10:124360.
25. Moreira AL, Ocampo PSS, Xia Y, Zhong H, Russell PA, Minami Y, et al. A grading
system for invasive pulmonary adenocarcinoma: a proposal from the Interna-
tional Association for the Study of Lung Cancer Pathology Committee. J Thorac
Oncol. 2020;15:1599610.
26. Kubouchi Y, Yurugi Y, Wakahara M, Sakabe T, Haruki T, Nosaka K, et al. Podo-
planin expression in cancer-associated broblasts predicts unfavourable prog-
nosis in patients with pathological stage IA lung adenocarcinoma.
Histopathology. 2018;72:4909.
27. Shi J, Hou Z, Yan J, Qiu W, Liang L, Meng M, et al. The prognostic signicance of
broblast activation protein-alpha in human lung adenocarcinoma. Ann Transl
Med. 2020;8:224.
28. Inoue C, Tamatsuki D, Miki Y, Saito R, Okada Y, Sasano H. Prognostic signicance
of combining immunohistochemical markers for cancer-associated broblasts in
lung adenocarcinoma tissue. Virchows Arch. 2019;475:1819.
29. Schalper KA, Brown J, Carvajal-Hausdorf D, McLaughlin J, Velcheti V, Syrigos KN,
et al. Objective measurement and clinical signicance of TILs in non-small cell
lung cancer. J Natl Cancer Inst. 2015;107:dju435.
30. Donnem T, Hald SM, Paulsen EE, Richardsen E, Al-Saad S, Kilvaer TK, et al. Stromal
CD8+T-cell densitya promising supplement to TNM staging in non-small cell
lung cancer. Clin Cancer Res. 2015;21:263543.
31. Geng Y, Shao Y, He W, Hu W, Xu Y, Chen J, et al. Prognostic role of tumor-
inltrating lymphocytes in lung cancer: a meta-analysis. Cell Physiol Biochem.
2015;37:156071.
32. Conway EM, Pikor LA, Kung SH, Hamilton MJ, Lam S, Lam WL, et al. Macrophages,
inammation, and lung cancer. Am J Respir Crit Care Med. 2016;193:11630.
33. Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T-cell
Inltration and PD-L1. Cancer Res. 2015;75:213945.
34. Lin Z, Gu J, Cui X, Huang L, Li S, Feng J, et al. Deciphering microenvironment of
NSCLC based on CD8+TIL density and PD-1/PD-L1 expression. J Cancer.
2019;10:21122.
35. Parra ER, Villalobos P, Behrens C, Jiang M, Pataer A, Swisher SG, et al. Effect of
neoadjuvant chemotherapy on the immune microenvironment in non-small cell
lung carcinomas as determined by multiplex immunouorescence and image
analysis approaches. J Immunother Cancer. 2018;6:48.
36. Peng H, Wu X, Zhong R, Yu T, Cai X, Liu J, et al. Proling tumor immune
microenvironment of non-small cell lung cancer using multiplex immuno-
uorescence. bioRxiv. 2021. https://doi.org/10.1101/2021.05.28.446005.
37. Tao Y, Li Y, Liang B. Comprehensive analysis of microenvironment-related genes
in lung adenocarcinoma. Future Oncol. 2020;16:182537.
38. Ojlert AK, Halvorsen AR, Nebdal D, Lund-Iversen M, Solberg S, Brustugun OT, et al.
The immune microenvironment in non-small cell lung cancer is predictive of
prognosis after surgery. Mol Oncol. 2019;13:116679.
39. Huang J, Li J, Zheng S, Lu Z, Che Y, Mao S, et al. Tumor microenvironment
characterization identies two lung adenocarcinoma subtypes with specic
immune and metabolic state. Cancer Sci. 2020;111:187686.
40. Ito M, Ishii G, Nagai K, Maeda R, Nakano Y, Ochiai A. Prognostic impact of cancer-
associated stromal cells in patients with stage I lung adenocarcinoma. Chest.
2012;142:1518.
41. Koucky V, Boucek J, Fialova A. Immunology of plasmacytoid dendritic cells in
solid tumors: a brief review. Cancers. 2019;11:470
42. Zhang S, Liu W, Hu B, Wang P, Lv X, Chen S, et al. Prognostic signicance of
tumor-inltrating natural killer cells in solid tumors: a systematic review and
meta-analysis. Front Immunol. 2020;11:1242.
43. Herbst RS, Giaccone G, de Marinis F, Reinmuth N, Vergnenegre A, Barrios CH, et al.
Atezolizumab for rst-line treatment of PD-L1-selected patients with NSCLC. N
Engl J Med. 2020;383:132839.
44. Qu Y, Wen J, Thomas G, Yang W, Prior W, He W, et al. Baseline frequency of
inammatory Cxcl9-expressing tumor-associated macrophages predicts response
to avelumab treatment. Cell Rep. 2020;32:107873.
45. House IG, Savas P, Lai J, Chen AXY, Oliver AJ, Teo ZL, et al. Macrophage-derived
CXCL9 and CXCL10 are required for antitumor immune responses following
immune checkpoint blockade. Clin Cancer Res. 2020;26:487504.
46. Jiang W, He Y, He W, Wu G, Zhou X, Sheng Q, et al. Exhausted CD8+T cells in the
tumor immune microenvironment: new pathways to therapy. Front Immunol.
2020;11:622509.
47. Sholl LM, Hirsch FR, Hwang D, Botling J, Lopez-Rios F, Bubendorf L, et al. The
promises and challenges of tumor mutation burden as an immunotherapy bio-
marker: a perspective from the International Association for the Study of Lung
Cancer Pathology Committee. J Thorac Oncol. 2020;15:140924.
48. Okazaki T, Tamai K, Shibuya R, Nakamura M, Mochizuki M, Yamaguchi K, et al.
Periostin is a negative prognostic factor and promotes cancer cell proliferation in
non-small cell lung cancer. Oncotarget. 2018;9:3118799.
P. Bischoff et al.
6757
Oncogene (2021) 40:6748 – 6758
Content courtesy of Springer Nature, terms of use apply. Rights reserved
49. Yang H, Jiang P, Liu D, Wang HQ, Deng Q, Niu X, et al. Matrix metalloproteinase
11 is a potential therapeutic target in lung adenocarcinoma. Mol Ther Oncolytics.
2019;14:8293.
50. Wrangle JM, Velcheti V, Patel MR, Garrett-Mayer E, Hill EG, Ravenel JG, et al. ALT-
803, an IL-15 superagonist, in combination with nivolumab in patients with
metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b
trial. Lancet Oncol. 2018;19:694704.
51. Patel AJ, Richter A, Drayson MT, Middleton GW. The role of B lymphocytes in the
immuno-biology of non-small-cell lung cancer. Cancer Immunol Immunother.
2020;69:32542.
52. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, et al.
Signatures of mutational processes in human cancer. Nature. 2013;500:41521.
53. Liu Y, Zhang J, Li L, Yin G, Zhang J, Zheng S, et al. Genomic heterogeneity of
multiple synchronous lung cancer. Nat Commun. 2016;7:13200.
54. Niemira M, Collin F, Szalkowska A, Bielska A, Chwialkowska K, Reszec J, et al.
Molecular signature of subtypes of non-small-cell lung cancer by large-scale
transcriptional proling: identication of key modules and genes by weighted
gene co-expression network analysis (WGCNA). Cancers. 2019;12:37.
55. Molina-Romero C, Rangel-Escareno C, Ortega-Gomez A, Alanis-Funes GJ, Aviles-
Salas A, Avila-Moreno F, et al. Differential gene expression proles according to
the Association for the Study of Lung Cancer/American Thoracic Society/Eur-
opean Respiratory Society histopathological classication in lung adenocarci-
noma subtypes. Hum Pathol. 2017;66:18899.
56. Hao Y, Baker D, Ten Dijke P. TGF-beta-mediated epithelial-mesenchymal transi-
tion and cancer metastasis. Int J Mol Sci. 2019;20:2767.
57. Tam SY, Wu VWC, Law HKW. Hypoxia-induced epithelial-mesenchymal transition
in cancers: HIF-1alpha and beyond. Front Oncol. 2020;10:486.
58. Lastwika KJ, Wilson W 3rd, Li QK, Norris J, Xu H, Ghazarian SR, et al. Control of PD-
L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small
cell lung cancer. Cancer Res. 2016;76:22738.
59. Gao Y, Yang J, Cai Y, Fu S, Zhang N, Fu X, et al. IFN-gamma-mediated inhibition of
lung cancer correlates with PD-L1 expression and is regulated by PI3K-AKT sig-
naling. Int J Cancer. 2018;143:93143.
60. Tan AC. Targeting the PI3K/Akt/mTOR pathway in non-small cell lung cancer
(NSCLC). Thorac Cancer. 2020;11:5118.
61. Mohrherr J, Haber M, Breitenecker K, Aigner P, Moritsch S, Voronin V, et al. JAK-
STAT inhibition impairs K-RAS-driven lung adenocarcinoma progression. Int J
Cancer. 2019;145:337688.
62. Stahl PL, Salmen F, Vickovic S, Lundmark A, Navarro JF, Magnusson J, et al.
Visualization and analysis of gene expression in tissue sections by spatial tran-
scriptomics. Science. 2016;353:7882.
63. Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay PK,
Swerdlow H, et al. Simultaneous epitope and transcriptome measurement in
single cells. Nat Methods. 2017;14:8658.
64. Buenrostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales ML, Snyder MP, et al.
Single-cell chromatin accessibility reveals principles of regulatory variation. Nat-
ure. 2015;523:48690.
65. Rajewsky N, Almouzni G, Gorski SA, Aerts S, Amit I, Bertero MG, et al. LifeTime and
improving European healthcare through cell-based interceptive medicine. Nat-
ure. 2020;587:377386.
66. Lin JR, Izar B, Wang S, Yapp C, Mei S, Shah PM, et al. Highly multiplexed
immunouorescence imaging of human tissues and tumors using t-CyCIF and
conventional optical microscopes. Elife. 2018;7:e31657.
67. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM 3rd, et al.
Comprehensive integration of single-cell data. Cell. 2019;177:1888902.e21.
68. Hafemeister C, Satija R. Normalization and variance stabilization of single-cell
RNA-seq data using regularized negative binomial regression. Genome Biol.
2019;20:296.
69. Habermann AC, Gutierrez AJ, Bui LT, Yahn SL, Winters NI, Calvi CL, et al. Single-cell
RNA sequencing reveals probrotic roles of distinct epithelial and mesenchymal
lineages in pulmonary brosis. Sci Adv. 2020;6:eaba1972.
70. Tata PR, Rajagopal J. Plasticity in the lung: making and breaking cell identity.
Development. 2017;144:75566.
71. Schubert M, Klinger B, Klunemann M, Sieber A, Uhlitz F, Sauer S, et al.
Perturbation-response genes reveal signaling footprints in cancer gene expres-
sion. Nat Commun. 2018;9:20.
72. Holland CH, Tanevski J, Perales-Paton J, Gleixner J, Kumar MP, Mereu E, et al.
Robustness and applicability of transcription factor and pathway analysis tools on
single-cell RNA-seq data. Genome Biol. 2020;21:36.
73. Efremova M, Vento-Tormo M, Teichmann SA, Vento-Tormo R. CellPhoneDB:
inferring cell-cell communication from combined expression of multi-subunit
ligand-receptor complexes. Nat Protoc. 2020;15:1484506.
74. Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, et al. TCGAbiolinks:
an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids
Res. 2016;44:e71.
75. Barbie DA, Tamayo P, Boehm JS, Kim SY, Moody SE, Dunn IF, et al. Systematic RNA
interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature.
2009;462:10812.
76. Hanzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for
microarray and RNA-seq data. BMC Bioinform. 2013;14:7.
ACKNOWLEDGEMENTS
We thank Manuela Pacyna-Gengelbach and Barabara Meyer-Bartell for excellent
technical assistance. The results of this study are in part based upon data generated
by the TCGA Research Network: https://www.cancer.gov/tcga.
AUTHOR CONTRIBUTIONS
NB, FK and PB conceived and designed the study; PB, AT, FK, DH, AE, TD, JN and JR
contributed to clinical sample acquisition and preparation; AT, PB and XL conducted
experiments; PB, BO, JP, DB, NB, JW and FU performed bioinformatic analyses; PB, FK,
NB, PJ, CF, DH, MM and CS analyzed and interpreted data and/or supervised parts of
the study; PB wrote the paper; MM, FK and NB revised the paper; all authors provided
critical feedback and helped shaping the research, analysis and paper.
FUNDING
The work was in part funded by the Berlin Institute of Health (to PB, PJ, DH, MM, CS,
and NB), the German Cancer Consortium DKTK (to MM and NB), the German Research
Foundation DFG (RTG2424 CompCancer to NB) and the Einstein Foundation/Stiftung
Charite (to NB). PB is participant in the BIH-Charité Junior Clinician Scientist Program
funded by the CharitéUniversitätsmedizin Berlin and the Berlin Institute of Health.
PJ is participant in the BIH-Charité Digital Clinician Scientist Program funded by the
CharitéUniversitätsmedizin Berlin and the Berlin Institute of Health and the
German Research Foundation (DFG). Open Access funding enabled and organized by
Projekt DEAL.
COMPETING INTERESTS
The authors declare no competing interests.
ADDITIONAL INFORMATION
Supplementary information The online version contains supplementary material
available at https://doi.org/10.1038/s41388-021-02054-3.
Correspondence and requests for materials should be addressed to Philip Bischoff.
Reprints and permission information is available at http://www.nature.com/
reprints
Publishers note Springer Nature remains neutral with regard to jurisdictional claims
in published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made. The images or other third party
material in this article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
articles Creative Commons license and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly
from the copyright holder. To view a copy of this license, visit http://creativecommons.
org/licenses/by/4.0/.
© The Author(s) 2021
P. Bischoff et al.
6758
Oncogene (2021) 40:6748 – 6758
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... p < 0.001). These patterns were derived from scRNA-seq data comprising 114,489 high-quality single-cell transcriptomes, linking cellular diversity to patient outcomes [44]. ...
Article
Full-text available
Recent developments in single-cell multi-omics technologies have provided the ability to identify diverse cell types and decipher key components of the tumor microenvironment (TME), leading to important advancements toward a much deeper understanding of how tumor microenvironment heterogeneity contributes to cancer progression and therapeutic resistance. These technologies are able to integrate data from molecular genomic, transcriptomic, proteomics, and metabolomics studies of cells at a single-cell resolution scale that give rise to the full cellular and molecular complexity in the TME. Understanding the complex and sometimes reciprocal relationships among cancer cells, CAFs, immune cells, and ECs has led to novel insights into their immense heterogeneity in functions, which can have important consequences on tumor behavior. In-depth studies have uncovered immune evasion mechanisms, including the exhaustion of T cells and metabolic reprogramming in response to hypoxia from cancer cells. Single-cell multi-omics also revealed resistance mechanisms, such as stromal cell-secreted factors and physical barriers in the extracellular matrix. Future studies examining specific metabolic pathways and targeting approaches to reduce the heterogeneity in the TME will likely lead to better outcomes with immunotherapies, drug delivery, etc., for cancer treatments. Future studies will incorporate multi-omics data, spatial relationships in tumor micro-environments, and their translation into personalized cancer therapies. This review emphasizes how single-cell multi-omics can provide insights into the cellular and molecular heterogeneity of the TME, revealing immune evasion mechanisms, metabolic reprogramming, and stromal cell influences. These insights aim to guide the development of personalized and targeted cancer therapies, highlighting the role of TME diversity in shaping tumor behavior and treatment outcomes.
... Next, we looked at transcriptional differences between the ON and OFF groups through differential expression. Differentially expressed genes included cytokines and growth factors Ccl4, Cxcl2, Tgfb, and Cxcl14; extracellular matrix remodeling components Timp3, Col4a1, and Fbln5; and growth arrest-specific 6 (Gas6), which is upregulated during growth arrest (16,17) (Figure 3, E and F, and Supplemental Figure 3D). Given the driver mutation in our mouse model, we also looked at genes that are upregulated in fibroblasts in KRAS G12D -driven pancreatic cancer (18). ...
... Single-cell sequencing is a high-throughput technology that enables detailed analysis of epigenomic, transcriptomic, and genomic characteristics at the single-cell level. This technology has found broad applications across oncology, immunology, neuroscience, and other fields [9][10][11]. Due to the structural and functional complexity of brain cells, isolating individual cells for single-cell analysis remains challenging. ...
Article
Full-text available
Background The role of neurons in central post-stroke pain (CPSP) following thalamic hemorrhage remains unclear. This study aimed to identify key genes associated with post-thalamic hemorrhage pain and to explore their functions in neurons. Single-nucleus RNA sequencing (snRNA-seq) data from a mouse model was used for this analysis. Methods First, snRNA-seq data were analyzed to identify cell types associated with CPSP induced by thalamic hemorrhage. Differentially expressed genes (DEGs) in neurons were then screened between control and model groups, followed by the construction of a protein-protein interaction (PPI) network for the DEGs. CytoNCA was used to assess node connectivity in the PPI network, and the top 5 key genes were identified. Subsequently, transcription factor (TF)-mRNA and miRNA-mRNA networks were constructed, and small-molecule drugs potentially targeting these key genes were predicted. Finally, the expression differences of key genes in neurons were compared between the model and control groups. Results A total of 13 cell clusters were identified, categorized into 8 cell types: T cells, endothelial cells, monocytes, neural progenitor cells (NPCs), microglia, astrocytes, neurons, and oligodendrocytes. A total of 228 DEGs were detected in neurons when comparing the model group with the control group. The PPI network of the DEGs consisted of 126 nodes and 209 edges, identifying the top 5 key genes: Dlgap1, Cacna1c, Gria2, Hsp90ab1, and Gapdh. The miRNA-mRNA network included 68 miRNA-mRNA pairs, 62 miRNAs, and 5 mRNAs, while the TF-mRNA network consisted of 66 TF-mRNA pairs, 56 TFs, and 5 mRNAs. Drug prediction identified 110 small-molecule drugs (e.g., purpurogallin, nifedipine, and novobiocin) potentially targeting these key genes. Additionally, Cacna1c were significantly upregulated in model mice. Conclusion This study identified the role of key genes in thalamic hemorrhage-induced CPSP through snRNA-seq, providing a scientific basis for further exploration of the molecular mechanisms underlying CPSP.
... In our study, we incorporated 10 samples from the dataset by Philip Bischoff and colleagues [26], which comprised 5 samples from individuals with normal lung tissue and 5 from patients with Lung Adenocarcinoma (LUAD). We leveraged the Seurat R package for the analysis of single-cell RNA sequencing data. ...
Article
Full-text available
Background Efferocytosis (ER) refers to the process of phagocytic clearance of programmed dead cells, and studies have shown that it is closely related to tumor immune escape. Methods This study was based on a comprehensive analysis of TCGA, GEO and CTRP databases. ER-related genes were collected from previous literature, univariate Cox regression was performed and consistent clustering was performed to categorize lung adenocarcinoma (LUAD) patients into two subgroups. Lasso regression and multivariate Cox regression analyses were used to construct ER-related prognostic features, and multiple immune infiltration algorithms were used to assess the correlation between the extracellular burial-related risk score (ERGRS) and tumor microenvironment (TME). And the key gene HAVCR1 was identified by deep learning, etc. Finally, pan-cancer analysis of the key genes was performed and in vitro experiments were conducted to verify the promotional effect of HAVCR1 on LUAD progression. Results A total of 33 ER-related genes associated with the prognosis of LUAD were identified, and the prognostic signature of ERGRS was successfully constructed to predict the overall survival (OS) and treatment response of LUAD patients. The high-risk group was highly enriched in some oncogenic pathways, while the low-ERGRS group was highly enriched in some immune-related pathways. In addition, the high ERGRS group had higher TMB, TNB and TIDE scores and lower immune scores. The low-risk group had better immunotherapeutic response and less likelihood of immune escape. Drug sensitivity analysis revealed that BRD-K92856060, monensin and hexaminolevulinate may be potential therapeutic agents for the high-risk group. And ERGRS was validated in several cohorts. In addition, HAVCR1 is one of the key genes, and knockdown of HAVCR1 in vitro significantly reduced the proliferation, migration and invasion ability of lung adenocarcinoma cells. Conclusion Our study developed a novel prognostic signature of efferocytosis-related genes. This prognostic signature accurately predicted survival prognosis as well as treatment outcome in LUAD patients and explored the role of HAVCR1 in lung adenocarcinoma progression.
... The bulk RNA-seq data of LUAD used in this study from The Cancer Genome Atlas (TCGA) database. The single-cell RNA-seq data of LUAD collected from Bischoff etc. research (https://doi.org/10.24433/CO.0121060.v1.) [17]. We identi ed 522 samples of bulk RNA-seq data in this study, including 331 samples without lymphatic metastases (N0) and 187 samples with lymphatic metastases (N1, N2, N3). ...
Preprint
Full-text available
Objective Lung adenocarcinoma (LUAD) is the most prevalent histological subtype of lung cancer, and lymph node metastasis serves as a significant prognostic risk factor. The identification of molecular biomarkers associated with lymph node metastasis holds paramount importance in the prevention and treatment strategies for this condition. Methods We identified the GFBP1 as the biomarker with the highest risk for lymph node metastasis by bioinformatical analysis. And we conducted the cell proliferation, invasion, and migration assays in H1975 and H1299 cells by overexpressing IGFBP1. The single-cell-RNA-sequence data indicated that IGFBP1 facilitates the progression of LUAD cells through the MAPK signaling pathway. Subsequently, western blot analysis was performed to validate these findings, while the ERK inhibitor U0126 was employed for cellular experiments and in vivo verification to elucidate the precise biological function of IGFBP1. Results IGFBP1 emerged as the most prominent biomarker for lymph node metastasis risk. Difference was shown in immunohistochemistry, univariate and multivariate Cox regression analyses. The vitro experiments confirmed that the overexpression of IGFBP1 in H1299 and H1975 cells can significantly promote proliferation, migration and invasion capacities. Western blot analysis validated that IGFBP1 overexpression substantially increased p-ERK expression levels. The use of the ERK inhibitor U0126 in subcutaneous tumor formation demonstrated that U0126 effectively suppressed both proliferation and invasion in animal model. Conclusions IGFBP1 indicates the promotion of lymph node metastasis in LUAD by facilitating tumor proliferation, invasion, and migration through modulation of the MAPK-ERK signaling pathway. Targeting this pathway exhibits significant potential for inhibiting tumor progression.
Article
The expansion of single-cell analytical techniques has empowered the exploration of diverse biological questions at the individual cells. Droplet-based single-cell RNA sequencing (scRNA-seq) methods have been particularly widely used due to their high-throughput capabilities and small reaction volumes. While commercial systems have contributed to the widespread adoption of droplet-based scRNA-seq, their relatively high cost limits the ability to profile large numbers of cells and samples. Moreover, as the scale of single-cell sequencing continues to expand, accommodating diverse workflows and cost-effective multi-biospecimen profiling becomes more critical. Herein, we present inDrops-2, an open-source scRNA-seq technology designed to profile live or preserved cells with a sensitivity matching that of state-of-the-art commercial systems but at a 6-fold lower cost. We demonstrate the flexibility of inDrops-2, by implementing two prominent scRNA-seq protocols, based on exponential and linear amplification of barcoded-complementary DNA, and provide useful insights into the advantages and disadvantages inherent to each approach. We applied inDrops-2 to simultaneously profile multiple human lung carcinoma samples that had been subjected to cell preservation, long-term storage and multiplexing to obtain a multiregional cellular profile of the tumor microenvironment. The scalability, sensitivity and cost efficiency make inDrops-2 stand out among other droplet-based scRNA-seq methods, ideal for large-scale studies on rare cell molecular signatures.
Article
Purpose To detect the prognostic importance of liquid–liquid phase separation (LLPS) in lung adenocarcinoma. Methods The gene expression files, copy number variation data, and clinical data were downloaded from The Cancer Genome Atlas cohort. LLPS-related genes were acquired from the DrLLPS website. The prognostic model based on LLPS was constructed by the Cox regression and LASSO regression analyses after the identification of LLPS-related differentially expressed genes (DEGs). Gene Ontology functional and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed. The LLPS-related prognostic risk score was validated by GSE31210 and GSE72094. The overall survival of lung adenocarcinoma patients was predicted by plotting a nomogram. The biological features of the high-risk lung adenocarcinoma were evaluated by the CIBERSORT, ESTIMATE, Gene Set Variation Analysis, and Genomics of Drug Sensitivity in Cancer. Reverse transcription-quantitative polymerase chain reaction detected hub gene expression. Results A total of 91 DEGs were screened out in LLPS, among which 9 genes were discovered as prognostic biomarkers of lung adenocarcinoma. GRIA1, CRTAC1, MAGEA4, and MAPK4 were identified as hub genes by the LASSO Cox regression analysis. High-risk and low-risk groups were divided according to the risk index, with the high-risk group displaying a markedly worse outcome. CRTAC1 expression was significantly decreased, MAGEA4 and MAPK4 expressions were increased, while GRIA1 expression was altered in lung adenocarcinoma cells. Tumor microenvironment, signaling pathway enrichment, and drug sensitivity significantly differed between different risk groups. Conclusions This work proposed a prognostic tool based on the LLPS-related gene signature to offer prospective and effective biomarkers for lung adenocarcinoma prognosis.
Article
Full-text available
Background The lysosome plays a vitally crucial role in tumor development and is a major participant in the cell death process, involving aberrant functional and structural changes. However, there are few studies on lysosome-associated genes (LAGs) in lung adenocarcinoma (LUAD). Methods Bulk RNA-seq of LUAD was downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). The lysosome risk signature was constructed after univariate and least absolute shrinkage and selection operator (Lasso) cox regression analysis of the TCGA training set, and its capability was validated by additional validation sets from GEO. Single cell sequencing (scRNA) was obtained from GEO to analyze the differences of lysosome risk signature at the single-cell level and the differences in the function and pathway. In vitro experiments have validated the function of CTSH in LUAD. Results The risk signature contained seven key LAGs, and patients were categorized into high- and low-risk groups based on a specific calculation formula. The LAG risk signature, which accurately predicted the prognostic status of LUAD patients, was still regarded as an independent prognostic indicator in multifactorial cox regression analysis. Subsequently, the combination of the signature and key clinical information was used to construct a column-line diagram for clinical assessment, which had a high discriminatory power. Immune infiltration analysis from bulk RNA-seq and scRNA-seq indicated that the low-risk group was immune-activated and had a better benefit in the prediction of immunotherapy. Finally, we validated its role in inhibiting tumor proliferation and metastasis in LUAD cells by knockdown of CTSH. Conclusion We defined a new biomarker that provided unique insights for individualized survival prediction and immunotherapy recommendations for LUAD patients.
Article
Full-text available
Lung adenocarcinoma (LUAD) is a major contributor to cancer-related deaths, distinguished by its pronounced tumor heterogeneity and persistent challenges in overcoming drug resistance. In this study, we utilized single-cell RNA sequencing (scRNA-seq) to dissect the roles of programmed cell death (PCD) pathways, including apoptosis, necroptosis, pyroptosis, and ferroptosis, in shaping LUAD heterogeneity, immune infiltration, and prognosis. Among these, ferroptosis and pyroptosis were most significantly associated with favorable survival outcomes, highlighting their potential roles in enhancing anti-tumor immunity. Distinct PCD-related LUAD subtypes were identified, characterized by differential pathway activation and immune cell composition. Subtypes enriched with cytotoxic lymphocytes and dendritic cells demonstrated improved survival outcomes and increased potential responsiveness to immunotherapy. Drug sensitivity analysis revealed that these subtypes exhibited heightened sensitivity to targeted therapies and immune checkpoint inhibitors, suggesting opportunities for personalized treatment strategies. Our findings emphasize the interplay between PCD pathways and the tumor microenvironment, providing insights into the mechanisms underlying tumor drug resistance and immune evasion. By linking molecular and immune features to clinical outcomes, this study highlights the potential of targeting PCD pathways to enhance therapeutic efficacy and overcome resistance in LUAD. These results contribute to a growing framework for developing precise and adaptable cancer therapies tailored to specific tumor characteristics. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-024-01736-0.
Article
Full-text available
Single-cell sequencing technology provides apparent advantages in cell population heterogeneity, allowing individuals to better comprehend tissues and organs. Sequencing technology is currently moving beyond the standard transcriptome to the single-cell level, which is likely to bring new insights into the function of breast cells. In this study, we examine the primary cell types involved in breast development, as well as achievements in the study of scRNA-seq in the microenvironment, stressing the finding of novel cell subsets using single-cell approaches and analyzing the problems and solutions to scRNA-seq. Furthermore, we are excited about the field’s promising future.
Article
Full-text available
This study attempted to profile the tumor immune microenvironment (TIME) of non-small cell lung cancer (NSCLC) by multiplex immunofluorescence of 681 NSCLC cases. The number, density, and proportion of 26 types of immune cells in tumor nest and tumor stroma were evaluated, revealing some close interactions particularly between intrastromal neutrophils and intratumoral regulatory T cells (Treg) (r ² = 0.439, P < 0.001), intrastromal CD4+CD38+ T cells and CD20-positive B cells (r ² = 0.539, P < 0.001), and intratumoral CD8-positive T cells and M2 macrophages expressing PD-L1 (r ² = 0.339, P < 0.001). Three immune subtypes correlated with distinct immune characteristics were identified using the unsupervised consensus clustering approach. The immune-activated subtype had the longest disease-free survival (DFS) and demonstrated the highest infiltration of CD4-positive T cells, CD8-positive T cells, and CD20-positive B cells. The immune-defected subtype was rich in cancer stem cells and macrophages, and these patients had the worst prognosis. The immune-exempted subtype had the highest levels of neutrophils and Tregs. Intratumoral CD68-positive macrophages, M1 macrophages, and intrastromal CD4+ cells, CD4+FOXP3- cells, CD8+ cells, and PD-L1+ cells were further found to be the most robust prognostic biomarkers for DFS, which were used to construct and validate the immune-related risk score for risk stratification (high vs. median vs. low) and the prediction of 5-year DFS rates (23.2% vs. 37.9% vs. 43.1%, P < 0.001). In conclusion, the intricate and intrinsic structure of TIME in NSCLC was demonstrated, showing potency in subtyping and prognostication.
Article
Full-text available
Tumor-specific CD8⁺T cells are exposed to persistent antigenic stimulation which induces a dysfunctional state called “exhaustion.” Though functioning to limit damage caused by immune response, T cell exhaustion leads to attenuated effector function whereby cytotoxic CD8⁺T cells fail to control tumor progression in the late stage. This pathway is a dynamic process from activation to “progenitor exhaustion” through to “terminally exhaustion” with distinct properties. With the rapid development of immunotherapy via enhancing T cell function, new studies are dissecting the mechanisms and identifying specific biomarkers of dynamic differentiation during the process of exhaustion. Further, although immune checkpoint inhibitors (ICIs) have achieved great success in clinical practice, most patients still show limited efficacy to ICIs. The expansion and differentiation of progenitor exhausted T cells explained the success of ICIs while the depletion of the progenitor T cell pool and the transient effector function of terminally exhausted T cells accounted for the failure of immune monotherapy in the context of exorbitant tumor burden. Thus, combination strategies are urgent to be utilized based on the reduction of tumor burden or the expansion of the progenitor T cell pool. In this review, we aim to introduce the concept of homeostasis of the activated and exhausted status of CD8⁺T cells in the tumor immune microenvironment, and present recent findings on dynamic differentiation process during T cell exhaustion and the implications for combination strategies in immune therapy.
Article
Full-text available
Although single-cell RNA sequencing studies have begun to provide compendia of cell expression profiles1, 2, 3, 4, 5, 6, 7, 8–9, it has been difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here, using droplet- and plate-based single-cell RNA sequencing of approximately 75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, we create an extensive cell atlas of the human lung. We define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 out of 45 previously known cell types and 14 previously unknown ones. This comprehensive molecular atlas identifies the biochemical functions of lung cells and the transcription factors and markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signalling interactions and immune cell homing; and identifies cell types that are directly affected by lung disease genes and respiratory viruses. By comparing human and mouse data, we identified 17 molecular cell types that have been gained or lost during lung evolution and others with substantially altered expression profiles, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This atlas provides the molecular foundation for investigating how lung cell identities, functions and interactions are achieved in development and tissue engineering and altered in disease and evolution.
Article
Full-text available
LifeTime aims to track, understand and target human cells during the onset and progression of complex diseases and their response to therapy at single-cell resolution. This mission will be implemented through the development and integration of single-cell multi-omics and imaging, artificial intelligence and patient-derived experimental disease models during progression from health to disease. Analysis of such large molecular and clinical datasets will discover molecular mechanisms, create predictive computational models of disease progression, and reveal new drug targets and therapies. Timely detection and interception of disease embedded in an ethical and patient-centered vision will be achieved through interactions across academia, hospitals, patient-associations, health data management systems and industry. Applying this strategy to key medical challenges in cancer, neurological, infectious, chronic inflammatory and cardiovascular diseases at the single-cell level will usher in cell-based interceptive medicine in Europe over the next decade.
Article
Full-text available
Pulmonary fibrosis (PF) is a form of chronic lung disease characterized by pathologic epithelial remodeling and accumulation of extracellular matrix (ECM). To comprehensively define the cell types, mechanisms, and mediators driving fibrotic remodeling in lungs with PF, we performed single-cell RNA sequencing of single-cell suspensions from 10 nonfibrotic control and 20 PF lungs. Analysis of 114,396 cells identified 31 distinct cell subsets/states. We report that a remarkable shift in epithelial cell phenotypes occurs in the peripheral lung in PF and identify several previously unrecognized epithelial cell phenotypes, including a KRT5 ⁻ / KRT17 ⁺ pathologic, ECM-producing epithelial cell population that was highly enriched in PF lungs. Multiple fibroblast subtypes were observed to contribute to ECM expansion in a spatially discrete manner. Together, these data provide high-resolution insights into the complexity and plasticity of the distal lung epithelium in human disease and indicate a diversity of epithelial and mesenchymal cells contribute to pathologic lung fibrosis.
Article
Full-text available
The tumor microenvironment is rich with immune-suppressive macrophages that are associated with cancer progression and resistance to immune checkpoint therapy. Using pre-treatment tumor biopsies complemented with single-cell RNA sequencing (RNA-seq), we characterize intratumoral immune heterogeneity to unveil potential mechanisms of resistance to avelumab (anti-PD-L1). We identify a proinflammatory F480⁺MHCII⁺Ly6Clo macrophage population that is associated with response rather than resistance to avelumab. These macrophages are the primary source of the interferon-inducible chemokine Cxcl9, which facilitates the recruitment of protective Cxcr3⁺ T cells. Consequently, the efficacy of avelumab in mouse tumor models is dependent on Cxcr3 and Cxcl9, and baseline levels of Cxcl9 in patients treated with avelumab are associated with clinical response and overall survival. These data suggest that, within the broadly immune-suppressive macrophage compartment, a pro-inflammatory population exists that promotes responsiveness to PD-L1 blockade.
Article
Full-text available
Background: Tumor-infiltrating natural killer (NK) cells (TINKs) are crucial immune cells in tumor defense, and might be related to tumor prognosis. However, the results were discrepant among different studies. The present meta-analysis was performed to comprehensively assess the prognostic value of NK cell markers in solid tumor tissues. Methods: PubMed, Web of Science, and EMBASE were searched to identify original researches reporting the prognostic significance of TINKs in solid tumors. NK cell markers CD56, CD57, NKp30, and NKp46 were included in the analysis. The hazard ratios (HRs) and 95% confidence intervals (CIs) of pooled overall survival (OS), disease-free survival (DFS), metastasis-free survival (MFS), progression-free survival (PFS), and recurrence-free survival (RFS) were calculated by STATA software 14.0 to assess the prognostic significance. Results : Of the 56 included studies, there were 18 studies on CD56, 31 studies on CD57, 1 study on NKp30, and 7 studies on NKp46. High levels of CD56, CD57, NKp30, and NKp46 were significantly correlated with better OS of patients with solid malignancies (HR = 0.473, 95%CI: 0.315–0.710, p < 0.001; HR = 0.484, 95%CI: 0.380–0.616, p < 0.001; HR = 0.34, 95%CI: 0.14–0.80, p = 0.014; HR = 0.622, 95%CI: 0.470–0.821, p < 0.001, respectively). Our results also revealed that CD56, CD57, and NKp46 could act as independent prognostic predictors for favorable OS (HR = 0.372, 95%CI: 0.261–0.531, p < 0.001; HR = 0.525, 95%CI: 0.346–0.797, p = 0.003; HR = 0.559, 95%CI: 0.385–0.812, p = 0.002, respectively). Conclusions : Our results indicated that high levels of NK cell markers in solid tumor tissues could predict favorable prognosis for solid tumor patients.
Preprint
Purpose We attempt to profile the tumor immune microenvironment (TIME) of non-small cell lung cancer (NSCLC) by multiplex immunofluorescence (MIF). Experimental Design MIF test was performed on 681 NSCLC cases in our center between 2009 and 2011. The number, density, proportion and correlation of 26 types of immune cells in tumor nest and tumor stroma were evaluated. An unsupervised consensus clustering approach was utilized to identify robust clusters of patients. Immune-related risk score (IRRS) was constructed for prognosis prediction for disease-free survival (DFS). Results The landscape of TIME was illustrated, revealing some close interactions particularly between intrastromal neutrophils and intratumoral regulatory T cells (Treg) (r ² = 0.439, P < 0.001), intrastromal CD4+CD38+ T cells and intrastromal CD20-positive B cells (r ² = 0.539, P < 0.001), and intratumoral CD8-positive T cells and intratumoral M2 macrophages expressing PD-L1 (r ² = 0.339, P < 0.001). Three immune subtypes correlated with distinct immune characteristics and clinical outcomes were identified. The immune-activated subtype had the longest DFS and demonstrated the highest infiltration of CD4-positive T cells and CD20-positive B cells. The immune-defected subtype had the highest levels of cancer stem cells and macrophages. The immune-exempted subtype had the highest levels of neutrophils and Treg. The IRRS based on six robust prognostic biomarkers showed potential ability for risk stratification (high vs. median vs. low) and prediction of five-year DFS rates (43.1% vs. 37.9% vs. 23.2%, P<0.001). Conclusions Our study profiled the intricate and intrinsic structure of TIME in NSCLC, which showed potency in subtyping and prognostication. Translational Relevance Significant unmet need exists in understanding the tumor immune microenvironment (TIME) of non-small cell lung cancer (NSCLC) and its correlation with prognosis. In this retrospective cohort study (n = 681), we profiled the immune landscape of NSCLC in situ and identified a novel stratification of TIME by three immune subtypes: immune-activated, immune-exempted, and immune-defected using multiplex immunofluorescence for testing 26 kinds of immune cells. Each of the immune subtypes was correlated with distinct composition, spatial distribution, and functional orientation of immune cells, and accordingly indicating significantly different disease-free survival (DFS). Close interactions were observed for several kinds of immune cells, including neutrophils and regulatory T cells, CD4+CD38+ T cells and CD20-positive B cells, and CD8-positive T cells and M2 macrophages. We also developed the immune-related risk score (IRRS) with different immune characteristics based on six robust immune biomarkers in TIME and evaluated its role in risk stratification and prognosis prediction of DFS. This study might bring potential clinical implementations for the design of novel immunotherapies and the optimization of combined strategies.
Article
Background: The efficacy and safety of the anti-programmed death ligand 1 (PD-L1) monoclonal antibody atezolizumab, as compared with those of platinum-based chemotherapy, as first-line treatment for patients with metastatic non-small-cell lung cancer (NSCLC) with PD-L1 expression are not known. Methods: We conducted a randomized, open-label, phase 3 trial involving patients with metastatic nonsquamous or squamous NSCLC who had not previously received chemotherapy and who had PD-L1 expression on at least 1% of tumor cells or at least 1% of tumor-infiltrating immune cells as assessed by the SP142 immunohistochemical assay. Patients were assigned in a 1:1 ratio to receive atezolizumab or chemotherapy. Overall survival (primary end point) was tested hierarchically according to PD-L1 expression status among patients in the intention-to-treat population whose tumors were wild-type with respect to EGFR mutations or ALK translocations. Within the population with EGFR and ALK wild-type tumors, overall survival and progression-free survival were also prospectively assessed in subgroups defined according to findings on two PD-L1 assays as well as by blood-based tumor mutational burden. Results: Overall, 572 patients were enrolled. In the subgroup of patients with EGFR and ALK wild-type tumors who had the highest expression of PD-L1 (205 patients), the median overall survival was longer by 7.1 months in the atezolizumab group than in the chemotherapy group (20.2 months vs. 13.1 months; hazard ratio for death, 0.59; P = 0.01). Among all the patients who could be evaluated for safety, adverse events occurred in 90.2% of the patients in the atezolizumab group and in 94.7% of those in the chemotherapy group; grade 3 or 4 adverse events occurred in 30.1% and 52.5% of the patients in the respective groups. Overall and progression-free survival favored atezolizumab in the subgroups with a high blood-based tumor mutational burden. Conclusions: Atezolizumab treatment resulted in significantly longer overall survival than platinum-based chemotherapy among patients with NSCLC with high PD-L1 expression, regardless of histologic type. (Funded by F. Hoffmann-La Roche/Genentech; IMpower110 ClinicalTrials.gov number, NCT02409342.).
Article
Introduction: A grading system for pulmonary adenocarcinoma has not been established. The International Association for the Study of Lung Cancer pathology panel evaluated a set of histologic criteria associated with prognosis aimed at establishing a grading system for invasive pulmonary adenocarcinoma. Methods: A multi-institutional study involving multiple cohorts of invasive pulmonary adenocarcinomas was conducted. A cohort of 284 stage I pulmonary adenocarcinomas was used as a training set to identify histologic features associated with patient outcomes (recurrence-free survival [RFS] and overall survival [OS]). Receiver operating characteristic curve analysis was used to select the best model, which was validated (n = 212) and tested (n = 300, including stage I-III) in independent cohorts. Reproducibility of the model was assessed using kappa statistics. Results: The best model (area under the receiver operating characteristic curve [AUC] = 0.749 for RFS and 0.787 for OS) was composed of a combination of predominant plus high-grade histologic pattern with a cutoff of 20% for the latter. The model consists of the following: grade 1, lepidic predominant tumor; grade 2, acinar or papillary predominant tumor, both with no or less than 20% of high-grade patterns; and grade 3, any tumor with 20% or more of high-grade patterns (solid, micropapillary, or complex gland). Similar results were seen in the validation (AUC = 0.732 for RFS and 0.787 for OS) and test cohorts (AUC = 0.690 for RFS and 0.743 for OS), confirming the predictive value of the model. Interobserver reproducibility revealed good agreement (k = 0.617). Conclusions: A grading system based on the predominant and high-grade patterns is practical and prognostic for invasive pulmonary adenocarcinoma.