Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell Rep ; 42(11): 113295, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37889752

ABSTRACT

Lung cancer treatment has benefited greatly through advancements in immunotherapies. However, immunotherapy often fails in patients with specific mutations like KEAP1, which are frequently found in lung adenocarcinoma. We established an antigenic lung cancer model and used it to explore how Keap1 mutations remodel the tumor immune microenvironment. Using single-cell technology and depletion studies, we demonstrate that Keap1-mutant tumors diminish dendritic cell and T cell responses driving immunotherapy resistance. This observation was corroborated in patient samples. CRISPR-Cas9-mediated gene targeting revealed that hyperactivation of the NRF2 antioxidant pathway is responsible for diminished immune responses in Keap1-mutant tumors. Importantly, we demonstrate that combining glutaminase inhibition with immune checkpoint blockade can reverse immunosuppression, making Keap1-mutant tumors susceptible to immunotherapy. Our study provides new insight into the role of KEAP1 mutations in immune evasion, paving the way for novel immune-based therapeutic strategies for KEAP1-mutant cancers.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Immune Evasion , Cell Line, Tumor , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/therapy , Adenocarcinoma of Lung/metabolism , Lung Neoplasms/therapy , Lung Neoplasms/drug therapy , Mutation/genetics , Immunotherapy , Tumor Microenvironment
2.
bioRxiv ; 2023 Mar 23.
Article in English | MEDLINE | ID: mdl-36993643

ABSTRACT

Tissue biology involves an intricate balance between cell-intrinsic processes and interactions between cells organized in specific spatial patterns, which can be respectively captured by single-cell profiling methods, such as single-cell RNA-seq (scRNA-seq), and histology imaging data, such as Hematoxylin-and-Eosin (H&E) stains. While single-cell profiles provide rich molecular information, they can be challenging to collect routinely and do not have spatial resolution. Conversely, histological H&E assays have been a cornerstone of tissue pathology for decades, but do not directly report on molecular details, although the observed structure they capture arises from molecules and cells. Here, we leverage adversarial machine learning to develop SCHAF (Single-Cell omics from Histology Analysis Framework), to generate a tissue sample's spatially-resolved single-cell omics dataset from its H&E histology image. We demonstrate SCHAF on two types of human tumors-from lung and metastatic breast cancer-training with matched samples analyzed by both sc/snRNA-seq and by H&E staining. SCHAF generated appropriate single-cell profiles from histology images in test data, related them spatially, and compared well to ground-truth scRNA-Seq, expert pathologist annotations, or direct MERFISH measurements. SCHAF opens the way to next-generation H&E2.0 analyses and an integrated understanding of cell and tissue biology in health and disease.

3.
J Clin Invest ; 133(9)2023 05 01.
Article in English | MEDLINE | ID: mdl-36951942

ABSTRACT

Patients with small cell lung cancer (SCLC) generally have a poor prognosis and a median overall survival of only about 13 months, indicating the urgent need for novel therapies. Delta-like protein 3 (DLL3) has been identified as a tumor-specific cell surface marker on neuroendocrine cancers, including SCLC. In this study, we developed a chimeric antigen receptor (CAR) against DLL3 that displays antitumor efficacy in xenograft and murine SCLC models. CAR T cell expression of the proinflammatory cytokine IL-18 greatly enhanced the potency of DLL3-targeting CAR T cell therapy. In a murine metastatic SCLC model, IL-18 production increased the activation of both CAR T cells and endogenous tumor-infiltrating lymphocytes. We also observed an increased infiltration, repolarization, and activation of antigen-presenting cells. Additionally, human IL-18-secreting anti-DLL3 CAR T cells showed an increased memory phenotype, less exhaustion, and induced durable responses in multiple SCLC models, an effect that could be further enhanced with anti-PD-1 blockade. All together, these results define DLL3-targeting CAR T cells that produce IL-18 as a potentially promising novel strategy against DLL3-expressing solid tumors.


Subject(s)
Carcinoma, Neuroendocrine , Interleukin-18 , Lung Neoplasms , Small Cell Lung Carcinoma , Animals , Humans , Mice , Cell Line, Tumor , Immunotherapy, Adoptive , Interleukin-18/pharmacology , Interleukin-18/therapeutic use , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , T-Lymphocytes/pathology , Xenograft Model Antitumor Assays
4.
Immunity ; 56(1): 93-106.e6, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36574773

ABSTRACT

Improved identification of anti-tumor T cells is needed to advance cancer immunotherapies. CD39 expression is a promising surrogate of tumor-reactive CD8+ T cells. Here, we comprehensively profiled CD39 expression in human lung cancer. CD39 expression enriched for CD8+ T cells with features of exhaustion, tumor reactivity, and clonal expansion. Flow cytometry of 440 lung cancer biospecimens revealed weak association between CD39+ CD8+ T cells and tumoral features, such as programmed death-ligand 1 (PD-L1), tumor mutation burden, and driver mutations. Immune checkpoint blockade (ICB), but not cytotoxic chemotherapy, increased intratumoral CD39+ CD8+ T cells. Higher baseline frequency of CD39+ CD8+ T cells conferred improved clinical outcomes from ICB therapy. Furthermore, a gene signature of CD39+ CD8+ T cells predicted benefit from ICB, but not chemotherapy, in a phase III clinical trial of non-small cell lung cancer. These findings highlight CD39 as a proxy of tumor-reactive CD8+ T cells in human lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Immune Checkpoint Inhibitors/therapeutic use , CD8-Positive T-Lymphocytes , Immunotherapy
5.
Cancer Res ; 82(3): 472-483, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34815254

ABSTRACT

Small cell lung cancer (SCLC) is an aggressive malignancy characterized by early metastasis and extreme lethality. The backbone of SCLC treatment over the past several decades has been platinum-based doublet chemotherapy, with the recent addition of immunotherapy providing modest benefits in a subset of patients. However, nearly all patients treated with systemic therapy quickly develop resistant disease, and there is an absence of effective therapies for recurrent and progressive disease. Here we conducted CRISPR-Cas9 screens using a druggable genome library in multiple SCLC cell lines representing distinct molecular subtypes. This screen nominated exportin-1, encoded by XPO1, as a therapeutic target. XPO1 was highly and ubiquitously expressed in SCLC relative to other lung cancer histologies and other tumor types. XPO1 knockout enhanced chemosensitivity, and exportin-1 inhibition demonstrated synergy with both first- and second-line chemotherapy. The small molecule exportin-1 inhibitor selinexor in combination with cisplatin or irinotecan dramatically inhibited tumor growth in chemonaïve and chemorelapsed SCLC patient-derived xenografts, respectively. Together these data identify exportin-1 as a promising therapeutic target in SCLC, with the potential to markedly augment the efficacy of cytotoxic agents commonly used in treating this disease. SIGNIFICANCE: CRISPR-Cas9 screening nominates exportin-1 as a therapeutic target in SCLC, and exportin-1 inhibition enhances chemotherapy efficacy in patient-derived xenografts, providing a novel therapeutic opportunity in this disease.


Subject(s)
Karyopherins/metabolism , Lung Neoplasms/drug therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Small Cell Lung Carcinoma/drug therapy , Animals , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Mice , Small Cell Lung Carcinoma/pathology , Exportin 1 Protein
6.
J Hematol Oncol ; 14(1): 170, 2021 10 16.
Article in English | MEDLINE | ID: mdl-34656143

ABSTRACT

BACKGROUND: Lineage plasticity, the ability to transdifferentiate among distinct phenotypic identities, facilitates therapeutic resistance in cancer. In lung adenocarcinomas (LUADs), this phenomenon includes small cell and squamous cell (LUSC) histologic transformation in the context of acquired resistance to targeted inhibition of driver mutations. LUAD-to-LUSC transdifferentiation, occurring in up to 9% of EGFR-mutant patients relapsed on osimertinib, is associated with notably poor prognosis. We hypothesized that multi-parameter profiling of the components of mixed histology (LUAD/LUSC) tumors could provide insight into factors licensing lineage plasticity between these histologies. METHODS: We performed genomic, epigenomics, transcriptomics and protein analyses of microdissected LUAD and LUSC components from mixed histology tumors, pre-/post-transformation tumors and reference non-transformed LUAD and LUSC samples. We validated our findings through genetic manipulation of preclinical models in vitro and in vivo and performed patient-derived xenograft (PDX) treatments to validate potential therapeutic targets in a LUAD PDX model acquiring LUSC features after osimertinib treatment. RESULTS: Our data suggest that LUSC transdifferentiation is primarily driven by transcriptional reprogramming rather than mutational events. We observed consistent relative upregulation of PI3K/AKT, MYC and PRC2 pathway genes. Concurrent activation of PI3K/AKT and MYC induced squamous features in EGFR-mutant LUAD preclinical models. Pharmacologic inhibition of EZH1/2 in combination with osimertinib prevented relapse with squamous-features in an EGFR-mutant patient-derived xenograft model, and inhibition of EZH1/2 or PI3K/AKT signaling re-sensitized resistant squamous-like tumors to osimertinib. CONCLUSIONS: Our findings provide the first comprehensive molecular characterization of LUSC transdifferentiation, suggesting putative drivers and potential therapeutic targets to constrain or prevent lineage plasticity.


Subject(s)
Adenocarcinoma of Lung/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Transdifferentiation , Humans , Mice, Inbred NOD , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction , Transcriptome
7.
Cancer Cell ; 39(11): 1479-1496.e18, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34653364

ABSTRACT

Small cell lung cancer (SCLC) is an aggressive malignancy that includes subtypes defined by differential expression of ASCL1, NEUROD1, and POU2F3 (SCLC-A, -N, and -P, respectively). To define the heterogeneity of tumors and their associated microenvironments across subtypes, we sequenced 155,098 transcriptomes from 21 human biospecimens, including 54,523 SCLC transcriptomes. We observe greater tumor diversity in SCLC than lung adenocarcinoma, driven by canonical, intermediate, and admixed subtypes. We discover a PLCG2-high SCLC phenotype with stem-like, pro-metastatic features that recurs across subtypes and predicts worse overall survival. SCLC exhibits greater immune sequestration and less immune infiltration than lung adenocarcinoma, and SCLC-N shows less immune infiltrate and greater T cell dysfunction than SCLC-A. We identify a profibrotic, immunosuppressive monocyte/macrophage population in SCLC tumors that is particularly associated with the recurrent, PLCG2-high subpopulation.


Subject(s)
Gene Expression Profiling/methods , Lung Neoplasms/genetics , Phospholipase C gamma/genetics , Small Cell Lung Carcinoma/genetics , Cell Plasticity , Humans , Neoplasm Metastasis , Prognosis , Sequence Analysis, RNA , Single-Cell Analysis , Survival Analysis
8.
iScience ; 24(11): 103224, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34712921

ABSTRACT

Activation of mitogenic signaling pathways is a common oncogenic driver of many solid tumors including lung cancer. Although activating mutations in the mitogen-activated protein kinase (MAPK) pathway are prevalent in non-small cell lung cancers, MAPK pathway activity, counterintuitively, is relatively suppressed in the more aggressively proliferative small cell lung cancer (SCLC). Here, we elucidate the role of the MAPK pathway and how it interacts with other signaling pathways in SCLC. We find that the most common SCLC subtype, SCLC-A associated with high expression of ASCL1, is selectively sensitive to MAPK activation in vitro and in vivo through induction of cell-cycle arrest and senescence. We show strong upregulation of ERK negative feedback regulators and STAT signaling upon MAPK activation in SCLC-A lines. These findings provide insight into the complexity of signaling networks in SCLC and suggest subtype-specific mitogenic vulnerabilities.

9.
Cancer Discov ; 11(12): 3214-3229, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34344693

ABSTRACT

Small cell lung cancer (SCLC) has limited therapeutic options and an exceptionally poor prognosis. Understanding the oncogenic drivers of SCLC may help define novel therapeutic targets. Recurrent genomic rearrangements have been identified in SCLC, most notably an in-frame gene fusion between RLF and MYCL found in up to 7% of the predominant ASCL1-expressing subtype. To explore the role of this fusion in oncogenesis and tumor progression, we used CRISPR/Cas9 somatic editing to generate a Rlf-Mycl-driven mouse model of SCLC. RLF-MYCL fusion accelerated transformation and proliferation of murine SCLC and increased metastatic dissemination and the diversity of metastatic sites. Tumors from the RLF-MYCL genetically engineered mouse model displayed gene expression similarities with human RLF-MYCL SCLC. Together, our studies support RLF-MYCL as the first demonstrated fusion oncogenic driver in SCLC and provide a new preclinical mouse model for the study of this subtype of SCLC. SIGNIFICANCE: The biological and therapeutic implications of gene fusions in SCLC, an aggressive metastatic lung cancer, are unknown. Our study investigates the functional significance of the in-frame RLF-MYCL gene fusion by developing a Rlf-Mycl-driven genetically engineered mouse model and defining the impact on tumor growth and metastasis. This article is highlighted in the In This Issue feature, p. 2945.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Gene Fusion , Genes, myc , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Proto-Oncogene Proteins c-myc , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Telomere-Binding Proteins
10.
Cancer Discov ; 11(12): 3028-3047, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34155000

ABSTRACT

Lineage plasticity is implicated in treatment resistance in multiple cancers. In lung adenocarcinomas (LUAD) amenable to targeted therapy, transformation to small cell lung cancer (SCLC) is a recognized resistance mechanism. Defining molecular mechanisms of neuroendocrine (NE) transformation in lung cancer has been limited by a paucity of pre/posttransformation clinical samples. Detailed genomic, epigenomic, transcriptomic, and protein characterization of combined LUAD/SCLC tumors, as well as pre/posttransformation samples, supports that NE transformation is primarily driven by transcriptional reprogramming rather than mutational events. We identify genomic contexts in which NE transformation is favored, including frequent loss of the 3p chromosome arm. We observed enhanced expression of genes involved in the PRC2 complex and PI3K/AKT and NOTCH pathways. Pharmacologic inhibition of the PI3K/AKT pathway delayed tumor growth and NE transformation in an EGFR-mutant patient-derived xenograft model. Our findings define a novel landscape of potential drivers and therapeutic vulnerabilities of NE transformation in lung cancer. SIGNIFICANCE: The difficulty in collection of transformation samples has precluded the performance of molecular analyses, and thus little is known about the lineage plasticity mechanisms leading to LUAD-to-SCLC transformation. Here, we describe biological pathways dysregulated upon transformation and identify potential predictors and potential therapeutic vulnerabilities of NE transformation in the lung. See related commentary by Meador and Lovly, p. 2962. This article is highlighted in the In This Issue feature, p. 2945.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Neuroendocrine Tumors , Small Cell Lung Carcinoma , Adenocarcinoma of Lung/drug therapy , Humans , Lung Neoplasms/drug therapy , Mutation , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Phosphatidylinositol 3-Kinases/genetics , Small Cell Lung Carcinoma/pathology
11.
Nat Cell Biol ; 21(4): 511-521, 2019 04.
Article in English | MEDLINE | ID: mdl-30886344

ABSTRACT

Recent studies have revealed a role for macrophages and neutrophils in limiting chemotherapy efficacy; however, the mechanisms underlying the therapeutic benefit of myeloid-targeting agents in combination with chemotherapy are incompletely understood. Here, we show that targeting tumour-associated macrophages by colony-stimulating factor-1 receptor (CSF-1R) blockade in the K14cre;Cdh1F/F;Trp53F/F transgenic mouse model for breast cancer stimulates intratumoural type I interferon (IFN) signalling, which enhances the anticancer efficacy of platinum-based chemotherapeutics. Notably, anti-CSF-1R treatment also increased intratumoural expression of type I IFN-stimulated genes in patients with cancer, confirming that CSF-1R blockade is a powerful strategy to trigger an intratumoural type I IFN response. By inducing an inflamed, type I IFN-enriched tumour microenvironment and by further targeting immunosuppressive neutrophils during cisplatin therapy, antitumour immunity was activated in this poorly immunogenic breast cancer mouse model. These data illustrate the importance of breaching multiple layers of immunosuppression during cytotoxic therapy to successfully engage antitumour immunity in breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Interferon Type I/physiology , Mammary Neoplasms, Experimental/drug therapy , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Humans , Immunity, Innate/drug effects , Macrophages/drug effects , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/secondary , Mice , Mice, Knockout , Mice, Transgenic
12.
Oncoimmunology ; 6(8): e1334744, 2017.
Article in English | MEDLINE | ID: mdl-28919995

ABSTRACT

Patients with primary solid malignancies frequently exhibit signs of systemic inflammation. Notably, elevated levels of neutrophils and their associated soluble mediators are regularly observed in cancer patients, and correlate with reduced survival and increased metastasis formation. Recently, we demonstrated a mechanistic link between mammary tumor-induced IL17-producing γδ T cells, systemic expansion of immunosuppressive neutrophils and metastasis formation in a genetically engineered mouse model for invasive breast cancer. How tumors orchestrate this systemic inflammatory cascade to facilitate dissemination remains unclear. Here we show that activation of this cascade relies on CCL2-mediated induction of IL1ß in tumor-associated macrophages. In line with these findings, expression of CCL2 positively correlates with IL1Β and macrophage markers in human breast tumors. We demonstrate that blockade of CCL2 in mammary tumor-bearing mice results in reduced IL17 production by γδ T cells, decreased neutrophil expansion and enhanced CD8+ T cell activity. These results highlight a new role for CCL2 in facilitating the breast cancer-induced pro-metastatic systemic inflammatory γδ T cell - IL17 - neutrophil axis.

13.
Nature ; 522(7556): 345-348, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-25822788

ABSTRACT

Metastatic disease remains the primary cause of death for patients with breast cancer. The different steps of the metastatic cascade rely on reciprocal interactions between cancer cells and their microenvironment. Within this local microenvironment and in distant organs, immune cells and their mediators are known to facilitate metastasis formation. However, the precise contribution of tumour-induced systemic inflammation to metastasis and the mechanisms regulating systemic inflammation are poorly understood. Here we show that tumours maximize their chance of metastasizing by evoking a systemic inflammatory cascade in mouse models of spontaneous breast cancer metastasis. We mechanistically demonstrate that interleukin (IL)-1ß elicits IL-17 expression from gamma delta (γδ) T cells, resulting in systemic, granulocyte colony-stimulating factor (G-CSF)-dependent expansion and polarization of neutrophils in mice bearing mammary tumours. Tumour-induced neutrophils acquire the ability to suppress cytotoxic T lymphocytes carrying the CD8 antigen, which limit the establishment of metastases. Neutralization of IL-17 or G-CSF and absence of γδ T cells prevents neutrophil accumulation and downregulates the T-cell-suppressive phenotype of neutrophils. Moreover, the absence of γδ T cells or neutrophils profoundly reduces pulmonary and lymph node metastases without influencing primary tumour progression. Our data indicate that targeting this novel cancer-cell-initiated domino effect within the immune system--the γδ T cell/IL-17/neutrophil axis--represents a new strategy to inhibit metastatic disease.


Subject(s)
Breast Neoplasms/pathology , Interleukin-17/biosynthesis , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neutrophils/metabolism , T-Lymphocyte Subsets/metabolism , Animals , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Granulocyte Colony-Stimulating Factor/immunology , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-17/immunology , Interleukin-1beta/immunology , Lung/pathology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymphatic Metastasis/immunology , Lymphatic Metastasis/pathology , Lymphocyte Activation , Mice , Neutrophils/cytology , Neutrophils/immunology , Phenotype , T-Lymphocyte Subsets/immunology , Tumor Microenvironment
14.
Pain ; 156(8): 1424-1432, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25734987

ABSTRACT

Morphine and other opioid analgesics are potent pain-relieving agents routinely used for pain management in patients with cancer. However, these drugs have recently been associated with a worse relapse-free survival in patients with surgical cancer, thus suggesting that morphine adversely affects cancer progression and relapse. In this study, we evaluated the impact of morphine on breast cancer progression, metastatic dissemination, and outgrowth of minimal residual disease. Using preclinical mouse models for metastatic invasive lobular and HER2 breast cancer, we show that analgesic doses of morphine do not affect mammary tumor growth, angiogenesis, and the composition of tumor-infiltrating immune cells. Our studies further demonstrate that morphine, administered in the presence or absence of surgery-induced tissue damage, neither facilitates de novo metastatic dissemination nor promotes outgrowth of minimal residual disease after surgery. Together, these findings indicate that opioid analgesics can be used safely for perioperative pain management in patients with cancer and emphasize that current standards of "good clinical practice" should be maintained.


Subject(s)
Breast Neoplasms/complications , Breast Neoplasms/pathology , Mammary Glands, Animal/drug effects , Morphine/adverse effects , Pain/drug therapy , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Animals , Breast Neoplasms/psychology , Carcinoma, Lobular/pathology , Disease Models, Animal , Disease Progression , Female , Genes, erbB-2/genetics , Humans , Mammary Glands, Animal/pathology , Mice , Mice, Knockout , Morphine/administration & dosage , Neoplasm Invasiveness/pathology , Pain/etiology , Pain/psychology , Treatment Outcome
15.
Cancer Res ; 73(1): 353-63, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23151903

ABSTRACT

Metastatic disease accounts for more than 90% of cancer-related deaths, but the development of effective antimetastatic agents has been hampered by the paucity of clinically relevant preclinical models of human metastatic disease. Here, we report the development of a mouse model of spontaneous breast cancer metastasis, which recapitulates key events in its formation and clinical course. Specifically, using the conditional K14cre;Cdh1(F/F);Trp53(F/F) model of de novo mammary tumor formation, we orthotopically transplanted invasive lobular carcinoma (mILC) fragments into mammary glands of wild-type syngeneic hosts. Once primary tumors were established in recipient mice, we mimicked the clinical course of treatment by conducting a mastectomy. After surgery, recipient mice succumbed to widespread overt metastatic disease in lymph nodes, lungs, and gastrointestinal tract. Genomic profiling of paired mammary tumors and distant metastases showed that our model provides a unique tool to further explore the biology of metastatic disease. Neoadjuvant and adjuvant intervention studies using standard-of-care chemotherapeutics showed the value of this model in determining therapeutic agents that can target early- and late-stage metastatic disease. In obtaining a more accurate preclinical model of metastatic lobular breast cancer, our work offers advances supporting the development of more effective treatment strategies for metastatic disease.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Lobular/pathology , Mammary Neoplasms, Experimental/pathology , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Animals , Breast Neoplasms/genetics , Carcinoma, Lobular/genetics , Female , Immunohistochemistry , In Situ Hybridization , Mammary Neoplasms, Experimental/genetics , Mice , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neoplasm Transplantation
16.
J Pathol ; 228(3): 300-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22926799

ABSTRACT

The leucine-rich repeat-containing heterotrimeric guanine nucleotide-binding protein-coupled receptor 5 (LGR5) has been identified as a marker of cycling stem cells in several epithelial tissues, including small intestine, colon, stomach and hair follicle. To investigate whether LGR5 also marks mammary epithelial stem cells, we performed in situ lineage-tracing studies and mammary gland reconstitutions with LGR5-expressing mammary epithelial cells. Interestingly, the LGR5 progeny population in mammary epithelium switches from the luminal to the myoepithelial compartment during the first 12 days of postnatal development, likely reflecting local changes in Wnt signalling. Together, our findings point to a stage-specific contribution of LGR5-expressing cells to luminal and basal epithelial lineages during postnatal mammary gland development.


Subject(s)
Cell Lineage/physiology , Epithelial Cells/cytology , Mammary Glands, Animal/growth & development , Morphogenesis/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , Epithelial Cells/physiology , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/physiology , Mice , Mice, Knockout , Models, Animal , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/physiology , Wnt Proteins/physiology
18.
Circ Res ; 110(4): 578-87, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22247485

ABSTRACT

RATIONALE: The importance for Bmp signaling during embryonic stem cell differentiation into myocardial cells has been recognized. The question when and where Bmp signaling in vivo regulates myocardial differentiation has remained largely unanswered. OBJECTIVE: To identify when and where Bmp signaling regulates cardiogenic differentiation. METHODS AND RESULTS: Here we have observed that in zebrafish embryos, Bmp signaling is active in cardiac progenitor cells prior to their differentiation into cardiomyocytes. Bmp signaling is continuously required during somitogenesis within the anterior lateral plate mesoderm to induce myocardial differentiation. Surprisingly, Bmp signaling is actively repressed in differentiating myocardial cells. We identified the inhibitory Smad6a, which is expressed in the cardiac tissue, to be required to inhibit Bmp signaling and thereby promote expansion of the ventricular myocardium. CONCLUSION: Bmp signaling exerts opposing effects on myocardial differentiation in the embryo by promoting as well as inhibiting cardiac growth.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Embryonic Stem Cells/metabolism , Heart/embryology , Myocytes, Cardiac/metabolism , Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Animals , Animals, Genetically Modified , Body Patterning , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/genetics , Cell Proliferation , Gene Expression Regulation, Developmental , Genes, Reporter , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Mutation , Smad6 Protein/metabolism , T-Box Domain Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
19.
J Pathol ; 224(1): 56-66, 2011 May.
Article in English | MEDLINE | ID: mdl-21480230

ABSTRACT

The tumour-modulating effects of the endogenous adaptive immune system are rather paradoxical. Whereas some clinical and experimental observations offer compelling evidence for the existence of immunosurveillance, other studies have revealed promoting effects of the adaptive immune system on primary cancer development and metastatic disease. We examined the functional significance of the adaptive immune system as a regulator of spontaneous HER2(+) breast tumourigenesis and pulmonary metastasis formation, using the MMTV-NeuT mouse model in which mammary carcinogenesis is induced by transgenic expression of the activated HER2/neu oncogene. Although T and B lymphocytes infiltrate human and experimental HER2(+) breast tumours, genetic elimination of the adaptive immune system does not affect development of premalignant hyperplasias or primary breast cancers. In addition, we demonstrate that pulmonary metastasis formation in MMTV-NeuT mice is not dependent on the adaptive immune system. Thus, our findings reveal that spontaneous HER2-driven mammary tumourigenesis and metastasis formation are neither suppressed, nor altered by immunosurveillance mechanisms, nor promoted by the adaptive immune system.


Subject(s)
Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/immunology , Receptor, ErbB-2/metabolism , Adaptive Immunity/immunology , Animals , Cell Transformation, Neoplastic/immunology , Female , Humans , Immune Tolerance/immunology , Immunologic Surveillance/immunology , Lung Neoplasms/immunology , Lymphocyte Subsets/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Neoplasm Transplantation , Tumor Microenvironment/immunology
20.
J Pathol ; 220(1): 34-44, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19927317

ABSTRACT

The contribution of cancer cell-intrinsic and -extrinsic factors to metastatic breast cancer is still poorly understood, hampering development of novel therapeutic strategies that decrease breast cancer mortality. Cre/loxP-based conditional mouse models of breast cancer present unique opportunities to study sporadic tumour formation and progression in a controlled setting. Unfortunately, the generation of mouse strains carrying multiple mutant alleles needed for such studies is very time-consuming. Moreover, conditional mouse tumour models do not permit independent manipulation of tumour cell-intrinsic and -extrinsic factors. Although the latter can be achieved by cleared fat-pad transplantation of mouse mammary epithelial cells (MMECs) from tumour suppressor gene (TSG) knockouts into wild-type or mutant recipients, this procedure is not possible for mutations that cause embryonic lethality or preclude mammary gland development. Here we show that cleared fat-pad transplantations with MMECs isolated from K14cre;Cdh1(F/F); Trp53(F/F) mice expressing Cre recombinase under control of the cytokeratin-14 promoter and carrying conditional null alleles for p53 and E-cadherin (Cdh1) first resulted in the formation of phenotypically normal mammary glands, followed by the development of invasive metastatic mammary tumours. Tumour formation in the recipients mimicked tumour latency, spectrum, morphology, immunophenotype, and metastatic characteristics of the original mammary tumour model. This transplantation system, which can be expanded to other conditional TSG knockouts, permits independent genetic analysis of stromal factors and testing of additional cancer cell-intrinsic mutations that would otherwise be embryonic lethal or require intensive breeding.


Subject(s)
Cell Transformation, Neoplastic/pathology , Mammary Glands, Animal/transplantation , Mammary Neoplasms, Experimental/pathology , Adipose Tissue/transplantation , Animals , Cadherins/metabolism , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/transplantation , Female , Keratin-8/metabolism , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Stochastic Processes , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...