Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cancer Cell ; 2024 May 24.
Article in English | MEDLINE | ID: mdl-38821060

ABSTRACT

Tumor metastasis requires systemic remodeling of distant organ microenvironments that impacts immune cell phenotypes, population structure, and intercellular communication. However, our understanding of immune phenotypic dynamics in the metastatic niche remains incomplete. Here, we longitudinally assayed lung immune transcriptional profiles in the polyomavirus middle T antigen (PyMT) and 4T1 metastatic breast cancer models from primary tumorigenesis, through pre-metastatic niche formation, to the final stages of metastatic outgrowth at single-cell resolution. Computational analyses of these data revealed a TLR-NFκB inflammatory program enacted by both peripherally derived and tissue-resident myeloid cells that correlated with pre-metastatic niche formation and mirrored CD14+ "activated" myeloid cells in the primary tumor. Moreover, we observed that primary tumor and metastatic niche natural killer (NK) cells are differentially regulated in mice and human patient samples, with the metastatic niche featuring elevated cytotoxic NK cell proportions. Finally, we identified cell-type-specific dynamic regulation of IGF1 and CCL6 signaling during metastatic progression that represents anti-metastatic immunotherapy candidate pathways.

2.
Clin Exp Metastasis ; 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38315348

ABSTRACT

Lymph nodes (LNs) are principal orchestrators of the adaptive immune response, yet in the context of malignancy, they are typically the first sites of metastasis. When tumors spread to LNs, they alter the immune repertoire, ultimately reconditioning it in a manner that suppresses anti-tumor immunity and promotes further metastatic dissemination. Conversely, activation of anti-tumor immunity within LNs is essential for immunotherapy, suggesting clinical approaches to radiotherapy in LNs and lymphadenectomy may need to be reconsidered in the context of immune checkpoint blockade (ICB). Herein, we discuss our understanding of the immune remodeling that coincides with LN metastasis as well as recent clinical studies exploring neoadjuvant immunotherapy and the roles of LNs in treatment of solid organ malignancies.

3.
bioRxiv ; 2023 May 07.
Article in English | MEDLINE | ID: mdl-37205523

ABSTRACT

Tumor metastasis requires systemic remodeling of distant organ microenvironments which impacts immune cell phenotypes, population structure, and intercellular communication networks. However, our understanding of immune phenotypic dynamics in the metastatic niche remains incomplete. Here, we longitudinally assayed lung immune cell gene expression profiles in mice bearing PyMT-driven metastatic breast tumors from the onset of primary tumorigenesis, through formation of the pre-metastatic niche, to the final stages of metastatic outgrowth. Computational analysis of these data revealed an ordered series of immunological changes that correspond to metastatic progression. Specifically, we uncovered a TLR-NFκB myeloid inflammatory program which correlates with pre-metastatic niche formation and mirrors described signatures of CD14+ 'activated' MDSCs in the primary tumor. Moreover, we observed that cytotoxic NK cell proportions increased over time which illustrates how the PyMT lung metastatic niche is both inflammatory and immunosuppressive. Finally, we predicted metastasis-associated immune intercellular signaling interactions involving Igf1 and Ccl6 which may organize the metastatic niche. In summary, this work identifies novel immunological signatures of metastasis and discovers new details about established mechanisms that drive metastatic progression. In brief: McGinnis et al. report a longitudinal scRNA-seq atlas of lung immune cells in mice bearing PyMT-driven metastatic breast tumors and identify immune cell transcriptional states, shifts in population structure, and rewiring of cell-cell signaling networks which correlate with metastatic progression. Highlights: Longitudinal scRNA-seq reveals distinct stages of immune remodeling before, during, and after metastatic colonization in the lungs of PyMT mice.TLR-NFκB inflammation correlates with pre-metastatic niche formation and involves both tissue-resident and bone marrow-derived myeloid cell populations. Inflammatory lung myeloid cells mirror 'activated' primary tumor MDSCs, suggesting that primary tumor-derived cues induce Cd14 expression and TLR-NFκB inflammation in the lung. Lymphocytes contribute to the inflammatory and immunosuppressive lung metastatic microenvironment, highlighted by enrichment of cytotoxic NK cells in the lung over time. Cell-cell signaling network modeling predicts cell type-specific Ccl6 regulation and IGF1-IGF1R signaling between neutrophils and interstitial macrophages.

4.
Trends Cell Biol ; 33(12): 1021-1034, 2023 12.
Article in English | MEDLINE | ID: mdl-37149414

ABSTRACT

Metastasis to lymph nodes (LNs) is a common feature of disease progression in most solid organ malignancies. Consequently, LN biopsy and lymphadenectomy are common clinical practices, not only because of their diagnostic utility but also as a means of deterring further metastatic spread. LN metastases have the potential to seed additional tissues and can induce metastatic tolerance, a process by which tumor-specific immune tolerance in LNs promotes further disease progression. Nonetheless, phylogenetic studies have revealed that distant metastases are not necessarily derived from nodal metastases. Furthermore, immunotherapy efficacy is increasingly being attributed to initiation of systemic immune responses within LNs. We argue that lymphadenectomy and nodal irradiation should be approached with caution, particularly in patients receiving immunotherapy.


Subject(s)
Lymph Nodes , Neoplasms , Humans , Phylogeny , Lymph Nodes/pathology , Lymph Node Excision , Neoplasms/therapy , Neoplasms/pathology , Disease Progression
5.
Cancer Cell ; 41(2): 356-372.e10, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36706760

ABSTRACT

Despite their cytotoxic capacity, neutrophils are often co-opted by cancers to promote immunosuppression, tumor growth, and metastasis. Consequently, these cells have received little attention as potential cancer immunotherapeutic agents. Here, we demonstrate in mouse models that neutrophils can be harnessed to induce eradication of tumors and reduce metastatic seeding through the combined actions of tumor necrosis factor, CD40 agonist, and tumor-binding antibody. The same combination activates human neutrophils in vitro, enabling their lysis of human tumor cells. Mechanistically, this therapy induces rapid mobilization and tumor infiltration of neutrophils along with complement activation in tumors. Complement component C5a activates neutrophils to produce leukotriene B4, which stimulates reactive oxygen species production via xanthine oxidase, resulting in oxidative damage and T cell-independent clearance of multiple tumor types. These data establish neutrophils as potent anti-tumor immune mediators and define an inflammatory pathway that can be harnessed to drive neutrophil-mediated eradication of cancer.


Subject(s)
Antineoplastic Agents , Neoplasms , Mice , Animals , Humans , Neutrophils , Neoplasms/drug therapy , Neoplasms/metabolism , Leukotriene B4/metabolism , Leukotriene B4/pharmacology , Tumor Necrosis Factor-alpha/metabolism
6.
Nat Methods ; 19(6): 759-769, 2022 06.
Article in English | MEDLINE | ID: mdl-35654951

ABSTRACT

Advances in multiplexed in situ imaging are revealing important insights in spatial biology. However, cell type identification remains a major challenge in imaging analysis, with most existing methods involving substantial manual assessment and subjective decisions for thousands of cells. We developed an unsupervised machine learning algorithm, CELESTA, which identifies the cell type of each cell, individually, using the cell's marker expression profile and, when needed, its spatial information. We demonstrate the performance of CELESTA on multiplexed immunofluorescence images of colorectal cancer and head and neck squamous cell carcinoma (HNSCC). Using the cell types identified by CELESTA, we identify tissue architecture associated with lymph node metastasis in HNSCC, and validate our findings in an independent cohort. By coupling our spatial analysis with single-cell RNA-sequencing data on proximal sections of the same specimens, we identify cell-cell crosstalk associated with lymph node metastasis, demonstrating the power of CELESTA to facilitate identification of clinically relevant interactions.


Subject(s)
Head and Neck Neoplasms , Cohort Studies , Humans , Lymphatic Metastasis , Squamous Cell Carcinoma of Head and Neck
7.
Cell ; 185(11): 1924-1942.e23, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35525247

ABSTRACT

For many solid malignancies, lymph node (LN) involvement represents a harbinger of distant metastatic disease and, therefore, an important prognostic factor. Beyond its utility as a biomarker, whether and how LN metastasis plays an active role in shaping distant metastasis remains an open question. Here, we develop a syngeneic melanoma mouse model of LN metastasis to investigate how tumors spread to LNs and whether LN colonization influences metastasis to distant tissues. We show that an epigenetically instilled tumor-intrinsic interferon response program confers enhanced LN metastatic potential by enabling the evasion of NK cells and promoting LN colonization. LN metastases resist T cell-mediated cytotoxicity, induce antigen-specific regulatory T cells, and generate tumor-specific immune tolerance that subsequently facilitates distant tumor colonization. These effects extend to human cancers and other murine cancer models, implicating a conserved systemic mechanism by which malignancies spread to distant organs.


Subject(s)
Lymph Nodes , Melanoma , Animals , Immune Tolerance , Immunotherapy , Lymphatic Metastasis/pathology , Melanoma/pathology , Mice
8.
Ann Transl Med ; 9(3): 218, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33708845

ABSTRACT

BACKGROUND: Skip metastasis is a common lymph node metastatic pattern in non-small cell lung cancer (NSCLC). The relationship between skip metastasis and specific clinicopathologic factors and the prognostic value of skip metastasis are controversial. METHODS: A systematic search and analysis of skip metastasis in NSCLC was conducted in the databases of PubMed, EMBASE, and Web of Science up to Dec 2019. Summarized hazard ratio (HR), mean difference (MD), and odds ratio (OR) with associated 95% confidence intervals (CI) were evaluated to investigating the relationship between skip metastasis and overall survival (OS), disease-free survival (DFS), recurrence-free survival (RFS) and clinicopathological features in NSCLC. RESULTS: 29 studies with a total of 1,806 skip and 4,670 non-skip N2 patients were included. The upper lobe tumor showed a higher rate of skip metastasis compared with lower lobe one (RR =1.16, 95% CI: 1.00-1.34, P=0.044, I2=39.8%). The presence of skip metastasis correlated with superior overall survival (HR =0.74, 95% CI: 0.66-0.83, P<0.001, I2=48.2%) and DFS or RFS (HR =0.71, 95% CI: 0.61-0.84, P<0.001, I2=18.2%). Further subgroup analyses indicated similar results in articles that reported intrapulmonary lymph node dissection (HR =0.67, 95% CI: 0.57-0.77, P<0.001, I2=0). CONCLUSIONS: The results indicate that the presence of skip metastasis is associated with a marked increase in survival of NSCLC patients compared to patients with non-skip N2 metastasis. These results suggest that skip metastasis might be a distinct subgroup for purposes of N staging of NSCLC patients, and intrapulmonary lymph node assessment is needed.

9.
Elife ; 92020 07 13.
Article in English | MEDLINE | ID: mdl-32657757

ABSTRACT

Tumor immunology is undergoing a renaissance due to the recent profound clinical successes of tumor immunotherapy. These advances have coincided with an exponential growth in the development of -omics technologies. Armed with these technologies and their associated computational and modeling toolsets, systems biologists have turned their attention to tumor immunology in an effort to understand the precise nature and consequences of interactions between tumors and the immune system. Such interactions are inherently multivariate, spanning multiple time and size scales, cell types, and organ systems, rendering systems biology approaches particularly amenable to their interrogation. While in its infancy, the field of 'Cancer Systems Immunology' has already influenced our understanding of tumor immunology and immunotherapy. As the field matures, studies will move beyond descriptive characterizations toward functional investigations of the emergent behavior that govern tumor-immune responses. Thus, Cancer Systems Immunology holds incredible promise to advance our ability to fight this disease.


Subject(s)
Immune System/physiology , Immunity , Neoplasms/immunology , Systems Biology
10.
Cancer Res ; 80(10): 1942-1956, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32127354

ABSTRACT

The recent success of checkpoint blockade therapies has established immunotherapy as one of the most promising treatments for melanoma. Nonetheless, a complete curative response following immunotherapy is observed only in a fraction of patients. To identify what factors limit the efficacy of immunotherapies, we established mouse models that cease to respond to immunotherapies once their tumors exceed a certain stage. Analysis of the immune systems of the organisms revealed that the numbers of tumor-infiltrating dendritic cells (TIDC) drastically decreased with time. Further, in contrast to the current paradigm, once melanoma was established, TIDC did not migrate into sentinel lymph nodes. Instead, they underwent local cell death due to excessive phagocytosis of lysosomes. Importantly, TIDC were required to license the cytotoxic activity of tumor CD8+ T cells, and in their absence, T cells did not lyse melanoma cells. Our results offer a paradigm shift regarding the role of TIDC and a framework to increase the efficacy of immunotherapies. SIGNIFICANCE: This work redefines the role of monocyte-derived dendritic cells in melanoma and provides a novel strategy to increase the efficacy of T-cell-based immunotherapies in nonresponding individuals. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/10/1942/F1.large.jpg.


Subject(s)
Dendritic Cells/pathology , Drug Resistance, Neoplasm/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lysosomes , Melanoma/immunology , Animals , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Immunotherapy , Lymphocyte Activation/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL
11.
J Clin Invest ; 129(10): 4151-4164, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31449054

ABSTRACT

While a high frequency of Th1 cells in tumors is associated with improved cancer prognosis, this benefit has been attributed mainly to support of cytotoxic activity of CD8+ T cells. By attempting to potentiate antibody-driven immunity, we found a remarkable synergy between CD4+ T cells and tumor-binding antibodies. This surprising synergy was mediated by a small subset of tumor-infiltrating CD4+ T cells that express the high-affinity Fcγ receptor for IgG (FcγRI) in both mouse and human patients. These cells efficiently lyse tumor cells coated with antibodies through concomitant crosslinking of their T cell receptor (TCR) and FcγRI. By expressing FcγRI and its signaling chain in conventional CD4+ T cells, we successfully employed this mechanism to treat established solid cancers. Overall, this discovery sheds new light on the biology of this T cell subset, their function during tumor immunity, and the means to utilize their unique killing signals in immunotherapy.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Receptors, IgG/metabolism , Th1 Cells/classification , Th1 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immunotherapy, Adoptive , Male , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocyte Subsets/immunology
12.
Nature ; 565(7741): 573-574, 2019 01.
Article in English | MEDLINE | ID: mdl-30683936
13.
Cancer Res ; 77(15): 4158-4170, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28611041

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) after complete surgical resection is often followed by distant metastatic relapse for reasons that remain unclear. In this study, we investigated how the immune response at secondary sites affects tumor spread in murine models of metastatic PDAC. Early metastases were associated with dense networks of CD11b+CD11c+MHC-II+CD24+CD64lowF4/80low dendritic cells (DC), which developed from monocytes in response to tumor-released GM-CSF. These cells uniquely expressed MGL2 and PD-L2 in the metastatic microenvironment and preferentially induced the expansion of T regulatory cells (Treg) in vitro and in vivo Targeted depletion of this DC population in Mgl2DTR hosts activated cytotoxic lymphocytes, reduced Tregs, and inhibited metastasis development. Moreover, blocking PD-L2 selectively activated CD8 T cells at secondary sites and suppressed metastasis, suggesting that the DCs use this particular pathway to inhibit CD8 T-cell-mediated tumor immunity. Phenotypically similar DCs accumulated at primary and secondary sites in other models and in human PDAC. These studies suggest that a discrete DC subset both expands Tregs and suppresses CD8 T cells to establish an immunosuppressive microenvironment conducive to metastasis formation. Therapeutic strategies to block the accumulation and immunosuppressive activity of such cells may help prevent PDAC progression and metastatic relapse after surgical resection. Cancer Res; 77(15); 4158-70. ©2017 AACR.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Dendritic Cells/immunology , Neoplasm Invasiveness/immunology , Pancreatic Neoplasms/pathology , Tumor Escape/immunology , Animals , Carcinoma, Pancreatic Ductal/immunology , Disease Models, Animal , Flow Cytometry , Mice , Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/immunology
14.
Cell ; 168(3): 487-502.e15, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28111070

ABSTRACT

Immune responses involve coordination across cell types and tissues. However, studies in cancer immunotherapy have focused heavily on local immune responses in the tumor microenvironment. To investigate immune activity more broadly, we performed an organism-wide study in genetically engineered cancer models using mass cytometry. We analyzed immune responses in several tissues after immunotherapy by developing intuitive models for visualizing single-cell data with statistical inference. Immune activation was evident in the tumor and systemically shortly after effective therapy was administered. However, during tumor rejection, only peripheral immune cells sustained their proliferation. This systemic response was coordinated across tissues and required for tumor eradication in several immunotherapy models. An emergent population of peripheral CD4 T cells conferred protection against new tumors and was significantly expanded in patients responding to immunotherapy. These studies demonstrate the critical impact of systemic immune responses that drive tumor rejection.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocyte Subsets/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Bone Marrow/immunology , Cell Proliferation , Disease Models, Animal , Female , Humans , Immune Tolerance , Killer Cells, Natural/immunology , Lymphocyte Activation , Lymphoid Tissue/immunology , Male , Melanoma/immunology , Melanoma/therapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Tumor Microenvironment
15.
JCI Insight ; 1(18): e89020, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27812544

ABSTRACT

BM-derived DC (BMDC) are powerful antigen-presenting cells. When loaded with immune complexes (IC), consisting of tumor antigens bound to antitumor antibody, BMDC induce powerful antitumor immunity in mice. However, attempts to employ this strategy clinically with either tumor-associated DC (TADC) or monocyte-derived DC (MoDC) have been disappointing. To investigate the basis for this phenomenon, we compared the response of BMDC, TADC, and MoDC to tumor IgG-IC. Our findings revealed, in both mice and humans, that upon exposure to IgG-IC, BMDC internalized the IC, increased costimulatory molecule expression, and stimulated autologous T cells. In contrast, TADC and, surprisingly, MoDC remained inert upon contact with IC due to dysfunctional signaling following engagement of Fcγ receptors. Such dysfunction is associated with elevated levels of the Src homology region 2 domain-containing phosphatase-1 (SHP-1) and phosphatases regulating Akt activation. Indeed, concomitant inhibition of both SHP-1 and phosphatases that regulate Akt activation conferred upon TADC and MoDC the capacity to take up and process IC and induce antitumor immunity in vivo. This work identifies the molecular checkpoints that govern activation of MoDC and TADC and their capacity to elicit T cell immunity.


Subject(s)
Antigen-Antibody Complex/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Lung Neoplasms/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Humans , Lymphocyte Activation , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Neoplasm Recurrence, Local , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Tumor Microenvironment
16.
Immunity ; 45(3): 641-655, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27590114

ABSTRACT

Although all-trans-retinoic acid (atRA) is a key regulator of intestinal immunity, its role in colorectal cancer (CRC) is unknown. We found that mice with colitis-associated CRC had a marked deficiency in colonic atRA due to alterations in atRA metabolism mediated by microbiota-induced intestinal inflammation. Human ulcerative colitis (UC), UC-associated CRC, and sporadic CRC specimens have similar alterations in atRA metabolic enzymes, consistent with reduced colonic atRA. Inhibition of atRA signaling promoted tumorigenesis, whereas atRA supplementation reduced tumor burden. The benefit of atRA treatment was mediated by cytotoxic CD8(+) T cells, which were activated due to MHCI upregulation on tumor cells. Consistent with these findings, increased colonic expression of the atRA-catabolizing enzyme, CYP26A1, correlated with reduced frequencies of tumoral cytotoxic CD8(+) T cells and with worse disease prognosis in human CRC. These results reveal a mechanism by which microbiota drive colon carcinogenesis and highlight atRA metabolism as a therapeutic target for CRC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Microbiota/immunology , Tretinoin/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis/immunology , Colon/immunology , Colon/metabolism , Colorectal Neoplasms/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Retinoic Acid 4-Hydroxylase/metabolism , Signal Transduction/immunology , Up-Regulation/immunology
17.
Cancer Discov ; 5(2): 168-81, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25421439

ABSTRACT

UNLABELLED: Metastasis is the leading cause of cancer-associated deaths. Although dissemination of tumor cells likely occurs early in tumorigenesis, the constituents of the microenvironment play essential rate-limiting roles in determining whether these cells will form clinically relevant tumors. Recent studies have uncovered many molecular factors that contribute to the establishment of a protumorigenic metastatic niche. Here, we demonstrate that galectin-3, whose expression has clinical associations with advanced malignancy and poor outcome, contributes to metastatic niche formation by binding to carbohydrates on metastatic cells. We show that galectin-3 is expressed early during tumorigenesis by both CD11b(+)Gr-1(+) and CD11b(+)Ly-6C(hi) leukocytes. Tumors mobilize these myeloid populations through secretion of soluble factors, including IL6. We find that metastatic cancer cells exhibit elevated presentation of the oncofetal galectin-3 carbohydrate ligand, the Thomsen-Friedenreich antigen, on their surfaces as a result of altered C2GnT2 and St6GalNAc4 glycosyltransferase activity that inhibits further glycosylation of this carbohydrate motif and promotes metastasis. SIGNIFICANCE: Although clinical observations of elevated serum galectin-3 levels and altered glycosylation have been associated with malignancy, we identify novel roles for glycosyltransferases in promoting adhesion to galectins in the metastatic niche. This identification of a cytokine-leukocyte-glycosylation axis in metastasis provides mechanistic explanations for clinical associations between malignancy and aberrant glycosylation.


Subject(s)
Galectin 3/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , N-Acetylglucosaminyltransferases/metabolism , Sialyltransferases/metabolism , Animals , Disease Models, Animal , Female , Glycosylation , Humans , Lung Neoplasms/genetics , Mice , Mice, Inbred C57BL , Neoplasm Metastasis
18.
Nat Commun ; 3: 1122, 2012.
Article in English | MEDLINE | ID: mdl-23047680

ABSTRACT

Extracellular matrix interactions have essential roles in normal physiology and many pathological processes. Although the importance of extracellular matrix interactions in metastasis is well documented, systematic approaches to identify their roles in distinct stages of tumorigenesis have not been described. Here we report a novel-screening platform capable of measuring phenotypic responses to combinations of extracellular matrix molecules. Using a genetic mouse model of lung adenocarcinoma, we measure the extracellular matrix-dependent adhesion of tumour-derived cells. Hierarchical clustering of the adhesion profiles differentiates metastatic cell lines from primary tumour lines. Furthermore, we uncovered that metastatic cells selectively associate with fibronectin when in combination with galectin-3, galectin-8 or laminin. We show that these molecules correlate with human disease and that their interactions are mediated in part by α3ß1 integrin. Thus, our platform allowed us to interrogate interactions between metastatic cells and their microenvironments, and identified extracellular matrix and integrin interactions that could serve as therapeutic targets.


Subject(s)
Adenocarcinoma/metabolism , Extracellular Matrix/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Fibronectins/metabolism , Galectin 3/metabolism , Galectins/metabolism , Laminin/metabolism , Lung Neoplasms/pathology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...