BMB Reports 2024; 57(11): 484-489  https://doi.org/10.5483/BMBRep.2024-0015
Characterization of an orthotopic mouse transplant model reveals early changes in the tumor microenvironment of lung cancer
Minsu Na1,2 , Huiram Kang1,2 , Nayoung Kim1,2 , Areum Jo1,2 & Hae-Ock Lee1,2,*
1Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, 2Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
Correspondence to: Tel: +82-2-3147-8365; Fax: +82-2-532-6537; E-mail: haeocklee@catholic.ac.kr
Received: January 20, 2024; Revised: February 13, 2024; Accepted: March 23, 2024; Published online: May 21, 2024.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

cc This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
To understand the cellular and molecular dynamics in the early stages of lung cancer, we explored a mouse model of orthotopic tumor transplant created from the Lewis Lung Carcinoma (LLC) cell line. Employing single-cell RNA sequencing, we analyzed the cellular landscape during tumor engraftment, focusing particularly on LLC cells harboring the Kras G12C mutation. This allowed us to identify LLC tumor cells via the detection of mutant Kras transcripts and observe elevated levels of Myc and mesenchymal gene expression. Moreover, our study revealed significant alterations in the lung microenvironment, including the activation of tissue remodeling genes in fibroblasts and the downregulation of MHC class II genes in myeloid subsets. Additionally, T/NK cell subsets displayed more regulatory phenotypes, coupled with reduced proliferation in CD8+ T cells. Collectively, these findings enhance our understanding of lung cancer progression, particularly in a tumor microenvironment with low immunogenicity.
Keywords: Lewis lung carcinoma, Lung cancer, Orthotopic transplant, Single-cell RNA sequencing, Tumor microenvironment
INTRODUCTION

The tumor microenvironment fosters tumor cell survival and is a key player in tumor progression (1, 2). The recent development of single-cell RNA sequencing (scRNA-seq) has allowed a systemic view of the tumor microenvironment. Comparisons between tumor and normal tissues have revealed alterations in immune cells such as a decrease in NK cells and an increase in suppressive or regulatory T cells (3, 4). Tumor associated macrophages demonstrate high levels of heterogeneity (5), and have emerged as a target for cancer treatment (6). In most solid tumors, cancer associated fibroblasts, including myofibroblasts, populate the tumor microenvironment (7, 8). However, the timing and sequence of the events remain unclear due to the long period of tumor progression before diagnosis and sampling.

Mouse models serve as useful tools for understanding the cellular and molecular dynamics of human cancer (9). Even in mouse models, capturing the early stages of endogenous cancer can be challenging due to low penetrance and long latency (10). Transplant models using cell lines may offer a practical alternative. In our study, we used a lung orthotopic tumor transplant model using the Lewis lung carcinoma (LLC) mouse lung cancer cell line (11). The LLC cell line harbors a heterozygous Kras G12C mutation (12), a prevalent driver mutation in both human and mouse lung cancer (13). KRAS mutations cause tumor-promoting inflammation and immune escape (14), which allows this model to reflect immunosuppressive lung adenocarcinoma that requires novel therapeutic approaches. Moreover, we used a method of intracardiac injection to simulate a multi-organ metastatic scenario (15). Our focus is on lung transplants, aiming to characterize the array of cellular and molecular changes that occur during the early stages of tumor engraftment.

RESULTS

Overview of scRNA-seq on LLC orthotopic mouse transplant model

To characterize cellular alterations following LLC engraftment in the lung, we established an orthotopic transplant model using intracardiac injection (Fig. 1A) (16). We assessed the presence of LLC cells using an in vivo imaging system (IVIS) to measure luciferase activity and detected multi-organ signals (Supplementary Fig. 1A). The hematoxylin and eosin (H&E) staining of tissue sections demonstrated the formation of tumor masses in the lung and liver (Fig. 1B). Notably, LLC tumor cells fail to grow in brain parenchyma (17). Subsequently, we performed scRNA-seq on the lungs (Fig. 1A). In the IVIS images, we distinguished between tumor-negative (without luminescence) and tumor-positive (with luminescence) lungs. In flow cytometry data, we observed immune (CD45+), epithelial (CD326+) and endothelial (CD31+) cells, but failed to identify LLC tumor cells (Supplementary Fig. 1B). We anticipated that the scRNA-seq data would enable us to annotate diverse cell types and provide a more detailed characterization.

Cellular landscape analysis identifies LLC tumor cells with Myc and Kras G12C expression

As illustrated in Fig. 1A, scRNA-seq was conducted on a cell suspension obtained by the enzymatic dissociation of lung tissues. After quality control filtering based on the number of genes and the percentage of mitochondrial genes (Supplementary Fig. 2A), 5,001 cells from tumor-negative and 3,007 cells from tumor-positive samples were subjected to further analysis (Fig. 2A and Supplementary Fig. 2B). Data integration and clustering using the Seurat pipeline (18), resulted in the identification of 12 cell types within the uniform manifold approximation and projection (UMAP) space (Fig. 2A). These cell types included tentative LLC tumor cells, mesothelial cells, fibroblasts, endothelial cells, normal epithelial cells (including AT1, AT2, goblet/club, and ciliated cells), macrophages, monocytes, T/NK cells, and B cells, identified by canonical marker gene expression (Fig. 2B). The LLC tumor cell clusters were verified by the KRAS mutation in sequencing BAM files, as marked on the feature plot (Fig. 2C, upper panel). Sequence comparisons in the integrated genomics viewer (IGV) (Supplementary Fig. 2C) reflected the heterozygous Kras G12C mutation in the LLC cell line. Notably, the Myc gene was highly expressed in LLC tumor clusters (Fig. 2C, lower). The tumor-positive lung contained most of the LLC tumor and mesothelial cells (Fig. 2A, right). Other cell types, including normal epithelial cells, were more abundant in the tumor-negative lung. To estimate overall changes in the cellular landscape, we compared differentially expressed genes (DEGs) between the total cells from tumor-negative and tumor-positive lung samples (Fig. 2D). In the tumor-positive lung, top DEGs including Myc and Cxcl1 were those most enriched in the LLC tumor cell clusters. By comparison, Sftpc in the top DEGs of tumor-negative lung is a AT2-specific gene. These results are consistent with the clustering analysis, showing tumor or AT-2 cell enrichment in the tumor-positive or tumor-negative lungs, respectively. We validated expression levels of the Myc, Cxcl1, and Sftpc genes by qRT-PCR (Fig. 2E).

Sub-clustering analyses identify cell type-specific genes modulated during tumor engraftment

Next, we performed sub-clustering analyses for fibroblast, myeloid (monocyte and macrophage clusters), and T/NK cell populations, which were present abundantly in both tumor-negative and positive lungs. First, fibroblasts were subdivided into ten clusters (18 PCs, res 0.6) representing six distinct subsets: adventitial fibroblasts, lipo-fibroblasts, peribronchiolar fibroblasts, Adh7+ fibroblasts, pericytes, and smooth muscle cells (SMCs) based on canonical marker expression (Fig. 3A and Supplementary Fig. 3A) (19). Notably, adventitial fibroblasts (Pi16, Serfinf1) exhibited elevated Fbln2 and Timp1 gene expression in the tumor-positive lung (Fig. 3A, lower). Lipo-fibroblasts displayed more pronounced gene expression (Inmt, Limch1) in the tumor-negative lung, whereas SMCs expressed the Acta2 and Myh11 genes in the tumor-positive lung. In the proportion, lipo-fibroblasts comprised the largest portion, with a slightly higher ratio of adventitial fibroblasts and a lower ratio of pericytes in the tumor-positive lung compared to the tumor-negative lung (Supplementary Fig. 3A, right).

Second, the analysis of myeloid cells revealed the presence of seven distinct clusters (14 PCs, res 0.3) representing four types of macrophages (monocyte-derived, inflammatory, interstitial, and alveolar), two types of dendritic cells (CD209a+ and plasmacytoid DCs), and monocytes/neutrophils (Fig. 3B and Supplementary Fig. 3B) (20). Interstitial macrophages expressed higher levels of C1qa, C1qb, and C1qc genes in the tumor-negative lung and showed downregulation of MHC class II genes (H2-Aa, H2-Eb1) in the tumor positive lung (Fig. 3B, lower). In the cell counts per cluster, macrophages were the most abundant in both tumor-positive and tumor-negative lungs (Supplementary Fig. 3B, right).

Finally, the analysis of the T/NK subset revealed eight clusters (15 PCs, res 0.6) representing 6 subpopulations: CD4 T cells, including naïve-like and regulatory T cells (Tregs), CD8 T cells with naïve-like or cytotoxic signatures, proliferating cells mostly with CD8+ T cell identity, NK cells, innate lymphoid cells (ILC2), and mucosal-associated invariant T cells (MAIT) with canonical marker expression (Fig. 3C and Supplementary Fig. 3C) (21, 22). In the proliferating cell cluster, the expression of Ctla4 gene was more pronounced in the tumor-positive lung, whereas the expression of proliferating genes was eminent only in the tumor-negative lung (Fig. 3C, lower). Cytotoxic CD8 T cells and proliferating cells were found at lower proportions in the tumor-positive lung (Supplementary Fig. 3C, right).

Analysis of cellular interactions reveals molecular connections between the LLC tumor and lung microenvironment

To gain insight into the cellular and molecular interactions driving tumor engraftment, we performed a correlation analysis between tumor cell clusters and the constituents of the tumor microenvironment (Fig. 4A). The analysis revealed a high gene expression correlation between LLC tumor cell clusters 4 and 6. Furthermore, copy number variation (CNV) inference confirmed similar genomic patterns in these tumor clusters (Fig. 4B). Then, we conducted a CellPhoneDB analysis (23) to infer molecular interactions between various cell types. Visualized as a heatmap, the interaction count network suggested vigorous cellular interactions (Fig. 4C). With a specific focus on LLC tumor cells, we identified receptor-ligand pairs that could mediate interactions with the microenvironment (Fig. 4D). Among these potential molecular interactions, SPP1-CD44, SPP1-integrin-complex, FN1-integrin-complex, and COL-integrin-complex interactions were prominent between LLC tumor cells and other cell types. ICAM1-AREG and LAMP1-FAM3C interactions were significant between mesothelial, fibroblast, endothelial, or epithelial cells and LLC tumor cells. FAM3C-CLEC2D interaction was notable among fibroblast, endothelial, and immune cells. TYROBP-CD44, C5AR1-RPS19, and CD74-MIF interactions were most prominent between myeloid cells and LLC tumor cells (Fig. 4D). We confirmed MIF protein expression in the LLC tumor mass and CD74 in the surrounding cells (Fig. 4E), as well as reduced MHC class II expression in the macrophages from the tumor-positive sample (Fig. 4F). Collectively, these molecular alterations suggest strong tissue remodeling and immunosuppression activities, which play a crucial role in promoting tumor engraftment within the lung microenvironment. 

DISCUSSION

In this study, we used scRNA-seq to explore the genomic features of the tumor transplant and the associated lung microenvironment. While previous studies have described the lung orthotopic model (24, 25), ours is the first to report systemic assessment of the total cell populations covering the tumor-stromal-immune axis.

In the analysis, we identified the LLC cells as a distinct cluster in the tumor-positive sample. The cluster exhibited high expression levels of Myc, Cxcl1, and Rhox5 genes, which are associated with tumor progression (26-28), yet lacked the typical marker of lung adenocarcinoma NK2-related homeobox transcription factor Nkx2-1 (29). In both mouse lung cancer models and human patients, Nkx2-1 is often lost in metastatic stages (2, 12). Notably, the LLC cluster showed high expression levels of mesenchymal genes such as Vim, Col3a1, and Mif corroborating the high metastatic potential of LLC cells (30).

To investigate the stromal-immune axis facilitating LLC engraftment, we employed two approaches, DEG and cellular network analysis. In the DEG analysis, a subset of fibroblasts displayed upregulation of Fbln2 and Timp1, which are genes involved in tissue remodeling and repair (31, 32). On the other hand, myeloid subsets showed a downregulation of MHC class II genes, suggesting dysfunctional antigen presentation (33, 34). Moreover, T/NK cell subsets exhibited a more regulatory phenotype but reduced proliferation-associated gene expression in CD8+ T cells. These findings indicate that tissue remodeling and immune suppression play crucial roles in creating a tumor-permissive environment. In the cellular network analysis, we found that mesenchymal genes in the LLC tumor probably engage in robust molecular interaction with various stromal cells, via pathways such as SPP1-CD44, SPP1-integrin-complex, FN1-integrin-complex, and COL-integrin-complex. Many of these alterations and interactions align with reports in human cancer data (4, 7), underscoring their contributions to tissue remodeling and cancer progression (35-37). Additional validation of these cellular and molecular interactions will be achieved through further analyses using spatial transcriptomics (38).

In summary, our study has revealed that an LLC transplant triggers significant changes in the tumor microenvironment. These changes are marked by the upregulation of tissue remodeling and repair genes in the stroma and the downregulation of MHC class II genes in macrophages. Notably, there is a more regulatory, less proliferative phenotype in the T/NK cell compartment, suggesting impaired antigen presentation and reduced T cell infiltration. These insights are crucial for understanding and potentially treating low-immunogenic tumors, particularly in the early stages.

MATERIALS AND METHODS

Cell lines

The LL/2-Luc2 (CRL-1642-LUC2) Lewis lung carcinoma cell line was purchased from the American Type Culture Collection (ATCC, Manassas, VA). Cells were cultured in DMEM containing 10% FBS and 2 μg/ml blasticidin S hydrochloride in a 5% CO2 incubator at 37°C.

Orthotopic mouse tumor model

Female C57BL/6 mice were purchased from Orient Bio Inc (Seongnam, Republic of Korea). LL/2-Luc2 cells (1 × 104 to 1 × 105 in 100 μl phosphate-buffered saline) were injected into the left ventricle of 8- to 9-week-old mice (16). Mice were monitored 2 to 3 times a week starting on the 5th day. Each mouse received an intraperitoneal injection of 100 μl of IVISbrite D-luciferin (Perkin Elmer, Waltham, MA), and bioluminescence images were acquired 10 minutes later using IVIS Lumina XRMS (Perkin Elmer).

All procedures involving animal research were in accordance with the Laboratory Animals Welfare Act, the Guide for the Care and Use of Laboratory Animals, and the Guidelines and Policies for Rodent experiment provided by the Institutional Animal Care and Use Committee (IACUC) in the School of Medicine, The Catholic University of Korea (Approval number: CUMS-2021-0363-06).

Sample collection for scRNA-seq

The lung tissue was minced, dissociated for 40 minutes at 37°C using the enzymes D, R, and A in RPMI1640 medium of the Tumor Dissociation Kit, mouse (Miltenyi Biotec, Bergisch Gladbach, Germany), and filtered through a 70-μm filter. Red blood cells were removed using RBC lysis solution (QIAGEN, Hilden, Germany).

Flow cytometry

Cells were incubated with TruStain FcX anti-mouse CD16/32 antibody (BioLegend, San Diego, CA) for 10 min and then stained with primary antibodies (Biolegend) for 40 minutes at 4°C before data analysis by FACS-Canto (BD Biosciences, San Jose, CA) and FlowJo software v.10.10.0.

Antibodies for total cell profiling: APC anti-CD45 (clone 30-F11); PE anti-CD90.2 (53-2.1); PerCP-Cy5.5 anti-CD31 (390); BV421 anti-CD326 (G8.8).

Antibodies for MHC class II expression by macrophages: PE Anti-CD45 (30-F11); FITC Anti-F4/80 (BM8); APC Anti-MHC II (M5/114.15.2).

Single-cell RNA sequencing

Single cell suspensions were processed using Chromium Next GEM Single Cell V(D)J Reagent Kits v2 (10× Genomics, Pleasanton, CA) with a cell recovery target of 7,000 per library according to the manufacturer’s instructions. Libraries were sequenced on an Illumina Hiseq X for 5’ GEX library, with 100PE.

scRNA-seq data analysis

The data was processed according to the Cell Ranger pipeline (version 5.0.0, 10× Genomics) and aligned to the mouse reference genome (mm10-2020-A). Gene expression profiles and the annotation information of single cells were compiled using the Seurat package (version 4) (18). Cells with less than 1,000 and more than 5,000 unique features or more than 10% of mitochondrial genes were removed from the merged data. Filtered cells were normalized with the LogNormalize function and highly variable features were identified using the FindVariableFeatures function. For batch correction, we used the Harmony package and performed principal component analysis (PCA) with the RunPCA function. Dimensionality reduction and clustering using UMAP were performed using the RunUMAP, Findneighbors, and FindClusters functions with 9 PCs, resolution = 0.3. Subset analyses were performed using a similar method with PC and resolution specified in the main text.

Kras G12C mutation detection

For the detection of the Kras G12C mutation, we used a method recommended by 10X Genomics, which involves creating a BAM file filtered by a specific barcode list (39). The nucleotide substitution from C to A marking the amino acid codon change from glycine (GGT) to cysteine (TGT) in the 12th amino acid position was confirmed in the IGV.

Copy number variation inference

To distinguish LLC tumors, we used the chromosomal gene expression pattern to infer CNVs. We established a reference group comprising all cell types except for LLC tumors and then created an inferCNV object (https://github.com/broadinstitute/inferCNV). We predicted copy number alteration (CNA) with the following settings: cutoff = 0.1, HMM_type = ‘i6’, tumor_subcluster_partition_method = “leiden”.

Cell–cell interaction analysis

CellPhoneDB (version 4.1.0) was used to infer cell-cell interactions (23). The UMI count matrix assigned to all cell types was loaded as an input onto the CellPhoneDB. Mouse gene symbols were converted to human ensemble gene IDs using the biomaRt package (version 2.46.3). Cells expressing specific ligands and receptors were set to more than 25%.

Immunohistochemistry staining

Lung tissues were fixed in 10% neutral buffered formalin, embedded in paraffin, and cut at 4-μm thickness. IHC staining was performed with Anti-CD74 (ab289885 from Abcam, Cambridge, UK, 1:2,000) and Anti-c-Myc antibodies (ab32072, Abcam, 1:50), followed by ImmPRESS HRP Universal Antibody (Horse Anti-Mouse/Rabbit IgG) Polymer Detection Kit (Vector Laboratories). Signals were detected with diaminobenzidine (DAB) and counterstained with hematoxylin. Stained slides were observed using an automatic digital slide scanner (Pannoramic SCAN II, 3DHISTECH) and analyzed with CaseViewer Software.

Quantitative RT-PCR

Total RNA was extracted using RNeasy Mini Kit (QIAGEN). RNA concentration was determined using a NanoDrop 2000/2000c Spectrophotometer (Thermo Scientific). Quantitative RT-PCR (qRT-PCR) was performed using the SuperScript First-Strand Synthesis System (Invitrogen) and IQ Sybr Green SuperMix (BioRad). Primer Sequences: Sftpc forward 5’-GTCCTCGTTGTCGTGGTGATTG-3’, reverse 5’-AAGGTAGCGATGGTGTCTGCTC-3’; Myc forward 5’-TCGCTGCTGTCCTCCGAGTCC-3’, reverse 5’-GGTTTGCCTCTTCTCCACAGAC-3’; Cxcl1 forward 5’-TCCAGAGCTTGAAGGTGTTGCC-3’, reverse 5’-AACCAAGGGAGCTTCAGGGTCA-3’.

Data availability

Raw and processed scRNA-seq data have been deposited at the Gene Expression Omnibus (GEO) with accession number GSE246710.

ACKNOWLEDGEMENTS

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (2022R1A2C1091451, RS-2023-00220840).

CONFLICTS OF INTEREST

The authors have no conflicting interests.

FIGURES
Fig. 1. Overview of scRNA-seq on LLC orthotopic mouse transplant model. (A) Overview of scRNA-seq analysis. After intracardiac injection of LLC cell line, tumor negative and positive lung tissues were subjected to scRNA-seq. (B) H&E staining of lung, liver, and brain tissues. The figure contrasts normal lung and liver tissues with those from the experiment.
Fig. 2. Cellular landscape analysis identifies LLC tumor cells with Myc and Kras G12C expression. (A) UMAP plot displaying 8,008 cells color-coded by sample, across 16 clusters or cell types (from left in order). Proportion bar plot representing 12 major cell types by sample (right, upper) and a frequency bar plot for each cell type (right, lower). (B) Dot plot illustrating major gene expression by cell type. (C) Feature plot showing Kras G12C genotype (upper) and Myc gene expression (lower). (D) Volcano plot highlighting DEGs between tumor-negative and tumor-positive samples. The x-axis represents log2 fold change (log2FC), and the y-axis shows P-value (log2FC > |2| & P-value < 10e-6). (E) Validation of Myc, Cxcl1, and Sftpc gene expression by qRT-PCR. Signal levels in normal lung tissue were established as the baseline, and the relative quantities (ΔCt) of each gene in tumor-positive lung were calculated (upper). After 40 cycles, the final products were run on a 1.5% agarose gel (lower).
Fig. 3. Subset analyses identify cell type-specific genes modulated during tumor engraftment. (A) Fibroblast sub-clustering analysis. UMAP plot color-coded by 6 fibroblast subsets (upper). Dot plot illustrating canonical marker gene expression (lower). (B) Myeloid sub-clustering analysis. UMAP plot color-coded by 7 myeloid subsets (upper). Dot plot showing canonical marker gene expression (lower). (C) T/NK cell sub-clustering analysis. UMAP plot color-coded by 8 T/NK subsets (upper). Dot plot displaying canonical marker gene expression (lower). (A-C) Dot plots are annotated by cell type (top) and cluster (bottom), with a color scale from −1 to 2 to represent cluster average expression and a size scale for the percentage of expressing cells within the cluster. Blue color bars mark tumor-negative and orange indicate tumor-positive samples.
Fig. 4. Molecular interactions between the tumor and its microenvironment. (A) Correlation plots showing the similarity of LLC tumor clusters 4 and 6. (B) Heatmap depicting the CNA profiles of LLC tumor clusters, in comparison with those of normal reference cell types. (C) Heatmap displaying the quantity of interactions between each cell type. (D) Dot plots showing significant means of receptor-ligand pair gene expression between LLC tumor and other cell types in the tumor-positive sample, with the circle size denoting significance. (E) Representative IHC images featuring CD74 and MIF expression, with an arrow pointing MIF-stained tumor. (F) Flow cytometry analysis utilizing anti-CD45, anti-F4/80, and anti-MHC class II antibodies to confirm MHC class II expression in macrophages.
REFERENCES
  1. Junttila MR and de Sauvage FJ (2013) Influence of tumour micro-environment heterogeneity on therapeutic response. Nature 501, 346-354.
    Pubmed CrossRef
  2. Altorki NK, Markowitz GJ and Gao D et al (2019) The lung microenvironment: an important regulator of tumour growth and metastasis. Nat Rev Cancer 19, 9-31.
    Pubmed KoreaMed CrossRef
  3. Lee HO, Hong Y and Etlioglu HE et al (2020) Lineage-dependent gene expression programs influence the immune landscape of colorectal cancer. Nat Genet 52, 594-603.
    Pubmed CrossRef
  4. Kim N, Kim HK and Lee K et al (2020) Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat Commun 11, 2285.
    Pubmed KoreaMed CrossRef
  5. Eum HH, Kwon M and Ryu D et al (2020) Tumor-promoting macrophages prevail in malignant ascites of advanced gastric cancer. Exp Mol Med 52, 1976-1988.
    Pubmed KoreaMed CrossRef
  6. Pittet MJ, Michielin O and Migliorini D (2022) Clinical relevance of tumour-associated macrophages. Nat Rev Clin Oncol 19, 402-421.
    Pubmed CrossRef
  7. Luo H, Xia X and Huang LB et al (2022) Pan-cancer single-cell analysis reveals the heterogeneity and plasticity of cancer-associated fibroblasts in the tumor microenvironment. Nat Commun 13, 6619.
    Pubmed KoreaMed CrossRef
  8. Hanley CJ, Waise S and Ellis MJ et al (2023) Single-cell analysis reveals prognostic fibroblast subpopulations linked to molecular and immunological subtypes of lung cancer. Nat Commun 14, 387.
    Pubmed KoreaMed CrossRef
  9. Hirst GL and Balmain A (2004) Forty years of cancer modelling in the mouse. Eur J Cancer 40, 1974-1980.
    Pubmed CrossRef
  10. Frese KK and Tuveson DA (2007) Maximizing mouse cancer models. Nat Rev Cancer 7, 645-658.
    Pubmed CrossRef
  11. Bertram JS and Janik P (1980) Establishment of a cloned line of Lewis Lung Carcinoma cells adapted to cell culture. Cancer Lett 11, 63-73.
    Pubmed CrossRef
  12. Agalioti T, Giannou AD and Krontira AC et al (2017) Mutant KRAS promotes malignant pleural effusion formation. Nat Commun 8, 15205.
    Pubmed KoreaMed CrossRef
  13. Drosten M, Guerra C and Barbacid M (2018) Genetically engineered mouse models of k-ras-driven lung and pancreatic tumors: validation of therapeutic targets. Cold Spring Harb Perspect Med 8, a031542.
    Pubmed KoreaMed CrossRef
  14. Hamarsheh S, Gross O, Brummer T and Zeiser R (2020) Immune modulatory effects of oncogenic KRAS in cancer. Nat Commun 11, 5439.
    Pubmed KoreaMed CrossRef
  15. Paku S, Dome B, Toth R and Timar J (2000) Organ-specificity of the extravasation process: an ultrastructural study. Clin Exp Metastasis 18, 481-492.
    Pubmed CrossRef
  16. Campbell JP, Merkel AR, Masood-Campbell SK, Elefteriou F and Sterling JA (2012) Models of bone metastasis. J Vis Exp 67, e4260.
    Pubmed CrossRef
  17. Valiente M, Van Swearingen AED and Anders CK et al (2020) Brain metastasis cell lines panel: a public resource of organotropic cell lines. Cancer Res 80, 4314-4323.
    Pubmed KoreaMed CrossRef
  18. Slovin S, Carissimo A and Panariello F et al (2021) Single-cell rna sequencing analysis: a step-by-step overview. Methods Mol Biol 2284, 343-365.
    Pubmed CrossRef
  19. Mayr CH, Sengupta A and Asgharpour S et al (2024) Sfrp1 inhibits lung fibroblast invasion during transition to injury induced myofibroblasts. Eur Respir J 63, 2301326.
    Pubmed KoreaMed CrossRef
  20. Gibbings SL, Thomas SM and Atif SM et al (2017) Three unique interstitial macrophages in the murine lung at steady state. Am J Respir Cell Mol Biol 57, 66-76.
    Pubmed KoreaMed CrossRef
  21. Asaoka M, Kabata H and Fukunaga K (2022) Heterogeneity of ILC2s in the Lungs. Front Immunol 13, 918458.
    Pubmed KoreaMed CrossRef
  22. Guo M, Morley MP and Jiang C et al (2023) Guided construction of single cell reference for human and mouse lung. Nat Commun 14, 4566.
    Pubmed KoreaMed CrossRef
  23. Efremova M, Vento-Tormo M, Teichmann SA and Vento-Tormo R (2020) CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat Protoc 15, 1484-1506.
    Pubmed CrossRef
  24. Poczobutt JM, De S and Yadav VK et al (2016) Expression profiling of macrophages reveals multiple populations with distinct biological roles in an immunocompetent orthotopic model of lung cancer. J Immunol 196, 2847-2859.
    Pubmed KoreaMed CrossRef
  25. Liu P, Zhao L, Senovilla L, Kepp O and Kroemer G (2021) In vivo imaging of orthotopic lung cancer models in mice. Methods Mol Biol 2279, 199-212.
    Pubmed CrossRef
  26. Gabay M, Li Y and Felsher DW (2014) MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb Perspect Med 4, a014241.
    Pubmed KoreaMed CrossRef
  27. Yuan M, Zhu H, Xu J, Zheng Y, Cao X and Liu Q (2016) Tumor-derived cxcl1 promotes lung cancer growth via recruitment of tumor-associated neutrophils. J Immunol Res 2016, 6530410.
    Pubmed KoreaMed CrossRef
  28. Li Q, O'Malley ME, Bartlett DL and Guo ZS (2011) Homeobox gene Rhox5 is regulated by epigenetic mechanisms in cancer and stem cells and promotes cancer growth. Mol Cancer 10, 63.
    Pubmed KoreaMed CrossRef
  29. Kwon MC and Berns A (2013) Mouse models for lung cancer. Mol Oncol 7, 165-177.
    Pubmed KoreaMed CrossRef
  30. Brodt P (1986) Characterization of two highly metastatic variants of Lewis lung carcinoma with different organ specificities. Cancer Res 46, 2442-2448.
    Pubmed
  31. Sasaki T, Gohring W, Pan TC, Chu ML and Timpl R (1995) Binding of mouse and human fibulin-2 to extracellular matrix ligands. J Mol Biol 254, 892-899.
    Pubmed CrossRef
  32. Gardner J and Ghorpade A (2003) Tissue inhibitor of metalloproteinase (TIMP)-1: the TIMPed balance of matrix metalloproteinases in the central nervous system. J Neurosci Res 74, 801-806.
    Pubmed KoreaMed CrossRef
  33. Bandola-Simon J and Roche PA (2019) Dysfunction of antigen processing and presentation by dendritic cells in cancer. Mol Immunol 113, 31-37.
    Pubmed KoreaMed CrossRef
  34. Wang H, Zhao S and Zhang X et al (2019) Major histocompatibility complex class II molecule in non-small cell lung cancer diagnosis, prognosis and treatment. Onco Targets Ther 12, 7281-7288.
    Pubmed KoreaMed CrossRef
  35. Spada S, Tocci A, Di Modugno F and Nistico P (2021) Fibronectin as a multiregulatory molecule crucial in tumor matrisome: from structural and functional features to clinical practice in oncology. J Exp Clin Cancer Res 40, 102.
    Pubmed KoreaMed CrossRef
  36. Zhao K, Ma Z and Zhang W (2021) Comprehensive analysis to identify SPP1 as a prognostic biomarker in cervical cancer. Front Genet 12, 732822.
    Pubmed KoreaMed CrossRef
  37. Su CY, Li JQ and Zhang LL et al (2020) The biological functions and clinical applications of integrins in cancers. Front Pharmacol 11, 579068.
    Pubmed KoreaMed CrossRef
  38. Lee J, Yoo M and Choi J (2022) Recent advances in spatially resolved transcriptomics: challenges and opportunities. BMB Rep 55, 113-124.
    Pubmed KoreaMed CrossRef
  39. Sinjab A, Han G and Treekitkarnmongkol W et al (2021) Resolving the spatial and cellular architecture of lung adenocarcinoma by multiregion single-cell sequencing. Cancer Discov 11, 2506-2523.
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles
  • CrossRef (0)

Author ORCID Information

Funding Information

Collections

Services
Social Network Service

e-submission

Archives