Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
2.
Oncoimmunology ; 12(1): 2201147, 2023.
Article in English | MEDLINE | ID: mdl-37089449

ABSTRACT

The clinical successes of immune checkpoint blockade (ICB) in advanced cancer patients have recently spurred the clinical implementation of ICB in the neoadjuvant and perioperative setting. However, how neoadjuvant ICB therapy affects the systemic immune landscape and metastatic spread remains to be established. Tumors promote both local and systemic expansion of regulatory T cells (Tregs), which are key orchestrators of tumor-induced immunosuppression, contributing to immune evasion, tumor progression and metastasis. Tregs express inhibitory immune checkpoint molecules and thus may be unintended targets for ICB therapy counteracting its efficacy. Using ICB-refractory models of spontaneous primary and metastatic breast cancer that recapitulate the poor ICB response of breast cancer patients, we observed that combined anti-PD-1 and anti-CTLA-4 therapy inadvertently promotes proliferation and activation of Tregs in the tumor, tumor-draining lymph node and circulation. Also in breast cancer patients, Treg levels were elevated upon ICB. Depletion of Tregs during neoadjuvant ICB in tumor-bearing mice not only reshaped the intratumoral immune landscape into a state favorable for ICB response but also induced profound and persistent alterations in systemic immunity, characterized by elevated CD8+ T cells and NK cells and durable T cell activation that was maintained after treatment cessation. While depletion of Tregs in combination with neoadjuvant ICB did not inhibit primary tumor growth, it prolonged metastasis-related survival driven predominantly by CD8+ T cells. This study demonstrates that neoadjuvant ICB therapy of breast cancer can be empowered by simultaneous targeting of Tregs, extending metastasis-related survival, independent of a primary tumor response.


Subject(s)
Breast Neoplasms , Lymphocyte Activation , T-Lymphocytes, Regulatory , Humans , Breast Neoplasms/immunology , Breast Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Neoadjuvant Therapy , Immune Checkpoint Inhibitors/therapeutic use , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Neoplasm Metastasis , Animals , Mice , CD8-Positive T-Lymphocytes/immunology
3.
Cancer Cell ; 41(1): 106-123.e10, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36525971

ABSTRACT

Immune checkpoint blockade (ICB) has heralded a new era in cancer therapy. Research into the mechanisms underlying response to ICB has predominantly focused on T cells; however, effective immune responses require tightly regulated crosstalk between innate and adaptive immune cells. Here, we combine unbiased analysis of blood and tumors from metastatic breast cancer patients treated with ICB with mechanistic studies in mouse models of breast cancer. We observe an increase in systemic and intratumoral eosinophils in patients and mice responding to ICB treatment. Mechanistically, ICB increased IL-5 production by CD4+ T cells, stimulating elevated eosinophil production from the bone marrow, leading to systemic eosinophil expansion. Additional induction of IL-33 by ICB-cisplatin combination or recombinant IL-33 promotes intratumoral eosinophil infiltration and eosinophil-dependent CD8+ T cell activation to enhance ICB response. This work demonstrates the critical role of eosinophils in ICB response and provides proof-of-principle for eosinophil engagement to enhance ICB efficacy.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Mice , Animals , Immune Checkpoint Inhibitors/therapeutic use , Eosinophils/pathology , Interleukin-5/therapeutic use , Interleukin-33 , Neoplasms/drug therapy , CD8-Positive T-Lymphocytes , Antigen Presentation , CD4-Positive T-Lymphocytes/pathology
4.
Oncoimmunology ; 11(1): 2063225, 2022.
Article in English | MEDLINE | ID: mdl-35481289

ABSTRACT

While regulatory T cells (Tregs) and macrophages have been recognized as key orchestrators of cancer-associated immunosuppression, their cellular crosstalk within tumors has been poorly characterized. Here, using spontaneous models for breast cancer, we demonstrate that tumor-associated macrophages (TAMs) contribute to the intratumoral accumulation of Tregs by promoting the conversion of conventional CD4+ T cells (Tconvs) into Tregs. Mechanistically, two processes were identified that independently contribute to this process. While TAM-derived TGF-ß directly promotes the conversion of CD4+ Tconvs into Tregsin vitro, we additionally show that TAMs enhance PD-1 expression on CD4+ T cells. This indirectly contributes to the intratumoral accumulation of Tregs, as loss of PD-1 on CD4+ Tconvs abrogates intratumoral conversion of adoptively transferred CD4+ Tconvs into Tregs. Combined, this study provides insights into the complex immune cell crosstalk between CD4+ T cells and TAMs in the tumor microenvironment of breast cancer, and further highlights that therapeutic exploitation of macrophages may be an attractive immune intervention to limit the accumulation of Tregs in breast tumors.


Subject(s)
Breast Neoplasms , T-Lymphocytes, Regulatory , Female , Humans , Immune Tolerance , Programmed Cell Death 1 Receptor , Tumor Microenvironment , Tumor-Associated Macrophages
5.
Cell Rep ; 38(9): 110447, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35235800

ABSTRACT

Breast cancer is accompanied by systemic immunosuppression, which facilitates metastasis formation, but how this shapes organotropism of metastasis is poorly understood. Here, we investigate the impact of mammary tumorigenesis on regulatory T cells (Tregs) in distant organs and how this affects multi-organ metastatic disease. Using a preclinical mouse mammary tumor model that recapitulates human metastatic breast cancer, we observe systemic accumulation of activated, highly immunosuppressive Tregs during primary tumor growth. Tumor-educated Tregs show tissue-specific transcriptional rewiring in response to mammary tumorigenesis. This has functional consequences for organotropism of metastasis, as Treg depletion reduces metastasis to tumor-draining lymph nodes, but not to lungs. Mechanistically, we find that Tregs control natural killer (NK) cell activation in lymph nodes, thereby facilitating lymph node metastasis. In line, an increased Treg/NK cell ratio is observed in sentinel lymph nodes of breast cancer patients compared with healthy controls. This study highlights that immune regulation of metastatic disease is highly organ dependent.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/pathology , Carcinogenesis/pathology , Female , Humans , Killer Cells, Natural/pathology , Lymph Nodes , Lymphatic Metastasis/pathology , Mice
6.
Gut Microbes ; 14(1): 2035660, 2022.
Article in English | MEDLINE | ID: mdl-35188867

ABSTRACT

The gut microbiota strongly impacts the development of sporadic colorectal cancer (CRC), but it is largely unknown how the microbiota affects the pathogenesis of mismatch-repair-deficient CRC in the context of Lynch syndrome. In a mouse model for Lynch syndrome, we found a nearly complete loss of intestinal tumor development when animals were transferred from a conventional "open" animal facility to specific-pathogen-free (SPF) conditions. Using 16S sequencing we detected large changes in microbiota composition between the two facilities. Transcriptomic analyses of tumor-free intestinal tissues showed signs of strong intestinal inflammation in conventional mice. Whole exome sequencing of tumors developing in Msh2-Lynch mice revealed a much lower mutational load in the single SPF tumor than in tumors developing in conventional mice, suggesting reduced epithelial proliferation in SPF mice. Fecal microbiota transplantations with conventional feces altered the immune landscape and gut homeostasis, illustrated by increased gut length and elevated epithelial proliferation and migration. This was associated with drastic changes in microbiota composition, in particular increased relative abundances of different mucus-degrading taxa such as Desulfovibrio and Akkermansia, and increased bacterial-epithelial contact. Strikingly, transplantation of conventional microbiota increased microsatellite instability in untransformed intestinal epithelium of Msh2-Lynch mice, indicating that the composition of the microbiota influences the rate of mutagenesis in MSH2-deficient crypts.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Gastrointestinal Microbiome , Animals , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Disease Models, Animal , Mice , MutS Homolog 2 Protein/genetics , Mutagenesis , Mutagens
7.
Annu Rev Cancer Biol ; 5: 291-310, 2021 Mar.
Article in English | MEDLINE | ID: mdl-34632244

ABSTRACT

The microenvironment of breast cancer hosts a dynamic cross talk between diverse players of the immune system. While cytotoxic immune cells are equipped to control tumor growth and metastasis, tumor-corrupted immunosuppressive immune cells strive to impair effective immunity and promote tumor progression. Of these, regulatory T cells (Tregs), the gatekeepers of immune homeostasis, emerge as multifaceted players involved in breast cancer. Intriguingly, clinical observations suggest that blood and intratumoral Tregs can have strong prognostic value, dictated by breast cancer subtype. Accordingly, emerging preclinical evidence shows that Tregs occupy a central role in breast cancer initiation and progression and provide critical support to metastasis formation. Here, Tregs are not only important for immune escape but also promote tumor progression independent of their immune regulatory capacity. Combining insights into Treg biology with advances made across the rapidly growing field of immuno-oncology is expected to set the stage for the design of more effective immunotherapy strategies.

8.
Cancer Cell ; 39(3): 301-303, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33513347

ABSTRACT

Neutrophils can facilitate the metastatic spread of cancer; however, how neutrophils are activated at metastatic sites remains poorly understood. In this issue, Xiao et al. demonstrate that the protease cathepsin C, secreted by breast cancer cells, triggers neutrophils to form neutrophil extracellular traps in the metastatic niche, thereby promoting lung metastasis.


Subject(s)
Extracellular Traps , Lung Neoplasms , Humans , Neutrophils , Soil
9.
Methods Enzymol ; 632: 259-281, 2020.
Article in English | MEDLINE | ID: mdl-32000899

ABSTRACT

Regulatory T cells (Tregs) play a major role in establishing an immunosuppressive tumor microenvironment. In order to fully uncover their role and molecular regulation in tumor-bearing hosts, it is critical to combine phenotypical characterization with functional analyses. A standard method to determine the suppressive potential of Tregs is with an in vitro suppression assay, in which the impact of freshly isolated Tregs on T cell proliferation is assessed. The assay requires the isolation of substantial numbers of Tregs from tissues and tumors, which can be challenging due to low yield or cell damage during sample preparation. In this chapter, we discuss a flexible suppression assay which can be used to assess the suppressive potential of low numbers of murine Tregs, directly isolated from tumors. We describe methods for tissue preparation, flow cytometry-based sorting of Tregs and optimal conditions to perform a suppression assay, to obtain reliable and reproducible results.


Subject(s)
Flow Cytometry/methods , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation , Female , Fluorescent Antibody Technique/methods , Immune Tolerance , Lymphocyte Activation , Mammary Neoplasms, Animal/immunology , Mice , Tumor Microenvironment
10.
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
11.
J Clin Invest ; 127(7): 2777-2788, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28581441

ABSTRACT

NK cells are highly efficient at preventing cancer metastasis but are infrequently found in the core of primary tumors. Here, have we demonstrated that freshly isolated mouse and human NK cells express low levels of the endo-ß-D-glucuronidase heparanase that increase upon NK cell activation. Heparanase deficiency did not affect development, differentiation, or tissue localization of NK cells under steady-state conditions. However, mice lacking heparanase specifically in NK cells (Hpsefl/fl NKp46-iCre mice) were highly tumor prone when challenged with the carcinogen methylcholanthrene (MCA). Hpsefl/fl NKp46-iCre mice were also more susceptible to tumor growth than were their littermate controls when challenged with the established mouse lymphoma cell line RMA-S-RAE-1ß, which overexpresses the NK cell group 2D (NKG2D) ligand RAE-1ß, or when inoculated with metastatic melanoma, prostate carcinoma, or mammary carcinoma cell lines. NK cell invasion of primary tumors and recruitment to the site of metastasis were strictly dependent on the presence of heparanase. Cytokine and immune checkpoint blockade immunotherapy for metastases was compromised when NK cells lacked heparanase. Our data suggest that heparanase plays a critical role in NK cell invasion into tumors and thereby tumor progression and metastases. This should be considered when systemically treating cancer patients with heparanase inhibitors, since the potential adverse effect on NK cell infiltration might limit the antitumor activity of the inhibitors.


Subject(s)
Heparin Lyase/immunology , Immunologic Surveillance , Killer Cells, Natural/immunology , Neoplasms, Experimental/immunology , Animals , Cell Line, Tumor , Cytokines/genetics , Cytokines/immunology , Female , Heparin Lyase/genetics , Humans , Killer Cells, Natural/pathology , Male , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/immunology , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/immunology , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/immunology
12.
Oncoimmunology ; 6(2): e1267892, 2017.
Article in English | MEDLINE | ID: mdl-28344878

ABSTRACT

The cytokine-induced SH2-containing protein CIS belongs to the suppressor of cytokine signaling (SOCS) protein family. Here, we show the critical role of CIS in suppressing natural killer (NK) cell control of tumor initiation and metastasis. Cish-deficient mice were highly resistant to methylcholanthrene-induced sarcoma formation and protected from lung metastasis of B16F10 melanoma and RM-1 prostate carcinoma cells. In contrast, the growth of primary subcutaneous tumors, including those expressing the foreign antigen OVA, was unchanged in Cish-deficient mice. The combination of Cish deficiency and relevant targeted and immuno-therapies such as combined BRAF and MEK inhibitors, immune checkpoint blockade antibodies, IL-2 and type I interferon revealed further improved control of metastasis. The data clearly indicate that targeting CIS promotes NK cell antitumor functions and CIS holds great promise as a novel target in NK cell immunotherapy.

13.
Pathog Dis ; 74(5)2016 07.
Article in English | MEDLINE | ID: mdl-27129365

ABSTRACT

Streptococcus mutans is a Gram-positive bacterium involved in development to caries, the most common infectious disease of our time. Streptococcus mutans interacts with other microbes, like the fungus Candida albicans and both are commonly isolated from patients with caries. Since the role of C. albicans in caries remains unknown, our aim was to unravel this using an in vitro dual-species cariogenic oral biofilm model. Biofilms were grown for 24-72 h on glass cover slips or hydroxyapatite (HA) disks to mimic the surface of teeth. Medium pH, lactic acid production capacity and calcium release from HA disks were determined. All 24-h biofilms had external pH values below the critical pH of 5.5 where enamel dissolves. In contrast, 72-h dual-species biofilms had significantly higher pH (above the critical pH) and consequently decreased calcium release compared to single-species S. mutans biofilms. Counter intuitively, lactic acid production and growth of S. mutans were increased in 72-h dual-species biofilms. Candida albicans modulates the pH in dual-species biofilms to values above the critical pH where enamel dissolves. Our results suggest that C. albicans is not by definition a cariogenic microorganism; it could prevent caries by actively increasing pH preventing mineral loss.


Subject(s)
Biofilms , Candida albicans/physiology , Dental Caries/prevention & control , Microbial Interactions , Mouth/microbiology , Calcium/metabolism , Colony Count, Microbial , Humans , Hydrogen-Ion Concentration , Lactic Acid/biosynthesis , Streptococcus mutans/physiology
SELECTION OF CITATIONS
SEARCH DETAIL