Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Nat Commun ; 15(1): 2806, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561380

ABSTRACT

Although heterogeneity of FAP+ Cancer-Associated Fibroblasts (CAF) has been described in breast cancer, their plasticity and spatial distribution remain poorly understood. Here, we analyze trajectory inference, deconvolute spatial transcriptomics at single-cell level and perform functional assays to generate a high-resolution integrated map of breast cancer (BC), with a focus on inflammatory and myofibroblastic (iCAF/myCAF) FAP+ CAF clusters. We identify 10 spatially-organized FAP+ CAF-related cellular niches, called EcoCellTypes, which are differentially localized within tumors. Consistent with their spatial organization, cancer cells drive the transition of detoxification-associated iCAF (Detox-iCAF) towards immunosuppressive extracellular matrix (ECM)-producing myCAF (ECM-myCAF) via a DPP4- and YAP-dependent mechanism. In turn, ECM-myCAF polarize TREM2+ macrophages, regulatory NK and T cells to induce immunosuppressive EcoCellTypes, while Detox-iCAF are associated with FOLR2+ macrophages in an immuno-protective EcoCellType. FAP+ CAF subpopulations accumulate differently according to the invasive BC status and predict invasive recurrence of ductal carcinoma in situ (DCIS), which could help in identifying low-risk DCIS patients eligible for therapeutic de-escalation.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Carcinoma, Intraductal, Noninfiltrating , Folate Receptor 2 , Humans , Female , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Fibroblasts/pathology , Cancer-Associated Fibroblasts/pathology , Extracellular Matrix/pathology , Tumor Microenvironment
3.
Breast Cancer Res ; 22(1): 76, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32665033

ABSTRACT

BACKGROUND: Early luminal breast cancer (BC) represents 70% of newly diagnosed BC cases. Among them, small (under 2 cm) BC without lymph node metastasis (classified as T1N0) have been rarely studied, as their prognosis is generally favorable. Nevertheless, up to 5% of luminal T1N0 BC patients relapse with distant metastases that ultimately prove fatal. The aim of our work was to identify the mechanisms involved in metastatic recurrence in these patients. METHODS: Our study addresses the role that autonomous and non-autonomous tumor cell features play with regard to distant recurrence in early luminal BC patients. We created a cohort of T1N0 luminal BC patients (tumors between 0.5-2 cm without lymph node metastasis) with metastatic recurrence ("cases") and corresponding "controls" (without relapse) matched 1:1 on main prognostic factors: age, grade, and proliferation. We deciphered different characteristics of cancer cells and their tumor micro-environment (TME) by deep analyses using immunohistochemistry. We performed in vitro functional assays and highlighted a new mechanism of cooperation between cancer cells and one particular subset of cancer-associated fibroblasts (CAF). RESULTS: We found that specific TME features are indicative of relapse in early luminal BC. Indeed, quantitative histological analyses reveal that "cases" are characterized by significant accumulation of a particular CAF subset (CAF-S1) and decrease in CD4+ T lymphocytes, without any other association with immune cells. In multivariate analysis, TME features, in particular CAF-S1 enrichment, remain significantly associated with recurrence, thereby demonstrating their clinical relevance. Finally, by performing functional analyses, we demonstrated that CAF-S1 pro-metastatic activity is mediated by the CDH11/osteoblast cadherin, consistent with bones being a major site of metastases in luminal BC patients. CONCLUSIONS: This study shows that distant recurrence in T1N0 BC is strongly associated with the presence of CAF-S1 fibroblasts. Moreover, we identify CDH11 as a key player in CAF-S1-mediated pro-metastatic activity. This is independent of tumor cells and represents a new prognostic factor. These results could assist clinicians in identifying luminal BC patients with high risk of relapse. Targeted therapies against CAF-S1 using anti-FAP antibody or CDH11-targeting compounds might help in preventing relapse for such patients with activated stroma.


Subject(s)
Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cancer-Associated Fibroblasts/immunology , Carcinoma, Ductal, Breast/immunology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/therapy , Carcinoma, Lobular/immunology , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Carcinoma, Lobular/therapy , Case-Control Studies , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Survival Rate , Tumor Microenvironment/immunology
4.
Cancer Discov ; 10(9): 1330-1351, 2020 09.
Article in English | MEDLINE | ID: mdl-32434947

ABSTRACT

A subset of cancer-associated fibroblasts (FAP+/CAF-S1) mediates immunosuppression in breast cancers, but its heterogeneity and its impact on immunotherapy response remain unknown. Here, we identify 8 CAF-S1 clusters by analyzing more than 19,000 single CAF-S1 fibroblasts from breast cancer. We validate the five most abundant clusters by flow cytometry and in silico analyses in other cancer types, highlighting their relevance. Myofibroblasts from clusters 0 and 3, characterized by extracellular matrix proteins and TGFß signaling, respectively, are indicative of primary resistance to immunotherapies. Cluster 0/ecm-myCAF upregulates PD-1 and CTLA4 protein levels in regulatory T lymphocytes (Tregs), which, in turn, increases CAF-S1 cluster 3/TGFß-myCAF cellular content. Thus, our study highlights a positive feedback loop between specific CAF-S1 clusters and Tregs and uncovers their role in immunotherapy resistance. SIGNIFICANCE: Our work provides a significant advance in characterizing and understanding FAP+ CAF in cancer. We reached a high resolution at single-cell level, which enabled us to identify specific clusters associated with immunosuppression and immunotherapy resistance. Identification of cluster-specific signatures paves the way for therapeutic options in combination with immunotherapies.This article is highlighted in the In This Issue feature, p. 1241.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Immune Checkpoint Inhibitors/pharmacology , Neoplasms/drug therapy , Tumor Escape , Tumor Microenvironment/immunology , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Datasets as Topic , Drug Resistance, Neoplasm/immunology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/surgery , Primary Cell Culture , RNA-Seq , Single-Cell Analysis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
5.
Nat Commun ; 11(1): 404, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31964880

ABSTRACT

Although fibroblast heterogeneity is recognized in primary tumors, both its characterization in and its impact on metastases remain unknown. Here, combining flow cytometry, immunohistochemistry and RNA-sequencing on breast cancer samples, we identify four Cancer-Associated Fibroblast (CAF) subpopulations in metastatic lymph nodes (LN). Two myofibroblastic subsets, CAF-S1 and CAF-S4, accumulate in LN and correlate with cancer cell invasion. By developing functional assays on primary cultures, we demonstrate that these subsets promote metastasis through distinct functions. While CAF-S1 stimulate cancer cell migration and initiate an epithelial-to-mesenchymal transition through CXCL12 and TGFß pathways, highly contractile CAF-S4 induce cancer cell invasion in 3-dimensions via NOTCH signaling. Patients with high levels of CAFs, particularly CAF-S4, in LN at diagnosis are prone to develop late distant metastases. Our findings suggest that CAF subset accumulation in LN is a prognostic marker, suggesting that CAF subsets could be examined in axillary LN at diagnosis.


Subject(s)
Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Lymphatic Metastasis/pathology , Myofibroblasts/metabolism , Adult , Aged , Aged, 80 and over , Axilla , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Cancer-Associated Fibroblasts/pathology , Cell Proliferation , Cell Separation , Chemokine CXCL12/metabolism , Epithelial-Mesenchymal Transition , Female , Flow Cytometry , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Lymph Nodes/cytology , Lymph Nodes/pathology , Middle Aged , Myofibroblasts/pathology , Neoplasm Invasiveness/pathology , Primary Cell Culture , Prognosis , Progression-Free Survival , Receptors, Notch/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Tumor Microenvironment
6.
Cell Rep ; 25(13): 3884-3893.e3, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30590056

ABSTRACT

A major challenge in cancer research is the complexity of the tumor microenvironment, which includes the host immunological setting. Inspired by the emerging technology of organ-on-chip, we achieved 3D co-cultures in microfluidic devices (integrating four cell populations: cancer, immune, endothelial, and fibroblasts) to reconstitute ex vivo a human tumor ecosystem (HER2+ breast cancer). We visualized and quantified the complex dynamics of this tumor-on-chip, in the absence or in the presence of the drug trastuzumab (Herceptin), a targeted antibody therapy directed against the HER2 receptor. We uncovered the capacity of the drug trastuzumab to specifically promote long cancer-immune interactions (>50 min), recapitulating an anti-tumoral ADCC (antibody-dependent cell-mediated cytotoxicity) immune response. Cancer-associated fibroblasts (CAFs) antagonized the effects of trastuzumab. These observations constitute a proof of concept that tumors-on-chip are powerful platforms to study ex vivo immunocompetent tumor microenvironments, to characterize ecosystem-level drug responses, and to dissect the roles of stromal components.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/pathology , Immunocompetence/drug effects , Tumor Microenvironment/drug effects , Animals , Cancer-Associated Fibroblasts/drug effects , Cattle , Cell Communication/drug effects , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Neoplasm Invasiveness , Receptor, ErbB-2/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Trastuzumab/pharmacology
7.
Nat Commun ; 9(1): 1056, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29535360

ABSTRACT

High-grade serous ovarian cancers (HGSOC) have been subdivided into molecular subtypes. The mesenchymal HGSOC subgroup, defined by stromal-related gene signatures, is invariably associated with poor patient survival. We demonstrate that stroma exerts a key function in mesenchymal HGSOC. We highlight stromal heterogeneity in HGSOC by identifying four subsets of carcinoma-associated fibroblasts (CAF-S1-4). Mesenchymal HGSOC show high content in CAF-S1 fibroblasts, which exhibit immunosuppressive functions by increasing attraction, survival, and differentiation of CD25+FOXP3+ T lymphocytes. The beta isoform of the CXCL12 chemokine (CXCL12ß) specifically accumulates in the immunosuppressive CAF-S1 subset through a miR-141/200a dependent-mechanism. Moreover, CXCL12ß expression in CAF-S1 cells plays a crucial role in CAF-S1 immunosuppressive activity and is a reliable prognosis factor in HGSOC, in contrast to CXCL12α. Thus, our data highlight the differential regulation of the CXCL12α and CXCL12ß isoforms in HGSOC, and reveal a CXCL12ß-associated stromal heterogeneity and immunosuppressive environment in mesenchymal HGSOC.


Subject(s)
Chemokine CXCL12/metabolism , Fibroblasts/metabolism , MicroRNAs/physiology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Female , Fibroblasts/cytology , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/genetics , Ovarian Neoplasms/genetics
8.
Cancer Cell ; 33(3): 463-479.e10, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29455927

ABSTRACT

Carcinoma-associated fibroblasts (CAF) are key players in the tumor microenvironment. Here, we characterize four CAF subsets in breast cancer with distinct properties and levels of activation. Two myofibroblastic subsets (CAF-S1, CAF-S4) accumulate differentially in triple-negative breast cancers (TNBC). CAF-S1 fibroblasts promote an immunosuppressive environment through a multi-step mechanism. By secreting CXCL12, CAF-S1 attracts CD4+CD25+ T lymphocytes and retains them by OX40L, PD-L2, and JAM2. Moreover, CAF-S1 increases T lymphocyte survival and promotes their differentiation into CD25HighFOXP3High, through B7H3, CD73, and DPP4. Finally, in contrast to CAF-S4, CAF-S1 enhances the regulatory T cell capacity to inhibit T effector proliferation. These data are consistent with FOXP3+ T lymphocyte accumulation in CAF-S1-enriched TNBC and show how a CAF subset contributes to immunosuppression.


Subject(s)
Fibroblasts/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Breast Neoplasms/immunology , Cell Differentiation/physiology , Cell Proliferation/physiology , Forkhead Transcription Factors/immunology , Humans , Immune Tolerance/immunology , Lymphocyte Activation/physiology
9.
Oncotarget ; 6(7): 4615-32, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25474135

ABSTRACT

Stress has an emerging role in cancer and targeting stress-related ß-adrenergic receptors (AR) has been proposed as a potential therapeutic approach in melanoma. Here we report that ß3-AR expression correlates with melanoma aggressiveness. In addition, we highlight that ß3-AR expression is not only restricted to cancer cells, but it is also expressed in vivo in stromal, inflammatory and vascular cells of the melanoma microenvironment. Particularly, we demonstrated that ß3-AR can (i) instruct melanoma cells to respond to environmental stimuli, (ii) enhance melanoma cells response to stromal fibroblasts and macrophages, (iii) increase melanoma cell motility and (iv) induce stem-like traits. Noteworthy, ß3-AR activation in melanoma accessory cells drives stromal reactivity by inducing pro-inflammatory cytokines secretion and de novo angiogenesis, sustaining tumor growth and melanoma aggressiveness. ß3-ARs also play a mandatory role in the recruitment to tumor sites of circulating stromal cells precursors, in the differentiation of these cells towards different lineages, further favoring tumor inflammation, angiogenesis and ultimately melanoma malignancy. Our findings validate selective ß3-AR antagonists as potential promising anti-metastatic agents. These could be used to complement current therapeutic approaches for melanoma patients (e.g. propranolol) by targeting non-neoplastic stromal cells, hence reducing therapy resistance of melanoma.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/pathology , Norepinephrine/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Skin Neoplasms/pathology , Tumor Microenvironment/drug effects , Apoptosis , Cell Proliferation , Cells, Cultured , Disease Progression , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunoenzyme Techniques , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Melanoma/drug therapy , Melanoma/metabolism , Neovascularization, Pathologic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Adrenergic, beta-3/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Vascular Endothelial Growth Factor A , Melanoma, Cutaneous Malignant
SELECTION OF CITATIONS
SEARCH DETAIL
...