Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 457
Filter
Add more filters

Publication year range
1.
Cell ; 185(7): 1189-1207.e25, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35325594

ABSTRACT

Macrophage infiltration is a hallmark of solid cancers, and overall macrophage infiltration correlates with lower patient survival and resistance to therapy. Tumor-associated macrophages, however, are phenotypically and functionally heterogeneous. Specific subsets of tumor-associated macrophage might be endowed with distinct roles on cancer progression and antitumor immunity. Here, we identify a discrete population of FOLR2+ tissue-resident macrophages in healthy mammary gland and breast cancer primary tumors. FOLR2+ macrophages localize in perivascular areas in the tumor stroma, where they interact with CD8+ T cells. FOLR2+ macrophages efficiently prime effector CD8+ T cells ex vivo. The density of FOLR2+ macrophages in tumors positively correlates with better patient survival. This study highlights specific roles for tumor-associated macrophage subsets and paves the way for subset-targeted therapeutic interventions in macrophages-based cancer therapies.


Subject(s)
Breast Neoplasms , Macrophages , Breast/immunology , Breast Neoplasms/epidemiology , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes , Female , Folate Receptor 2 , Humans , Lymphocytes, Tumor-Infiltrating , Prognosis
2.
Nature ; 620(7972): 181-191, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37380767

ABSTRACT

The adult human breast is comprised of an intricate network of epithelial ducts and lobules that are embedded in connective and adipose tissue1-3. Although most previous studies have focused on the breast epithelial system4-6, many of the non-epithelial cell types remain understudied. Here we constructed the comprehensive Human Breast Cell Atlas (HBCA) at single-cell and spatial resolution. Our single-cell transcriptomics study profiled 714,331 cells from 126 women, and 117,346 nuclei from 20 women, identifying 12 major cell types and 58 biological cell states. These data reveal abundant perivascular, endothelial and immune cell populations, and highly diverse luminal epithelial cell states. Spatial mapping using four different technologies revealed an unexpectedly rich ecosystem of tissue-resident immune cells, as well as distinct molecular differences between ductal and lobular regions. Collectively, these data provide a reference of the adult normal breast tissue for studying mammary biology and diseases such as breast cancer.


Subject(s)
Breast , Gene Expression Profiling , Single-Cell Analysis , Adult , Female , Humans , Breast/cytology , Breast/immunology , Breast/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Endothelial Cells/classification , Endothelial Cells/metabolism , Epithelial Cells/classification , Epithelial Cells/metabolism , Genomics , Immunity
3.
J Mammary Gland Biol Neoplasia ; 26(3): 247-261, 2021 09.
Article in English | MEDLINE | ID: mdl-34341887

ABSTRACT

A majority of breast cancers (BC) are age-related and we seek to determine what cellular and molecular changes occur in breast tissue with age that make women more susceptible to cancer initiation. Immune-epithelial cell interactions are important during mammary gland development and the immune system plays an important role in BC progression. The composition of human immune cell populations is known to change in peripheral blood with age and in breast tissue during BC progression. Less is known about changes in immune populations in normal breast tissue and how their interactions with mammary epithelia change with age. We quantified densities of T cells, B cells, and macrophage subsets in pathologically normal breast tissue from 122 different women who ranged in age from 24 to 74 years old. Donor-matched peripheral blood from a subset of 20 donors was analyzed by flow cytometry. Tissue immune cell densities and localizations relative to the epithelium were quantified in situ with machine learning-based image analyses of multiplex immunohistochemistry-stained tissue sections. In situ results were corroborated with flow cytometry analyses of peri-epithelial immune cells from primary breast tissue preparations and transcriptome analyses of public data from bulk tissue reduction mammoplasties. Proportions of immune cell subsets in breast tissue and donor-matched peripheral blood were not correlated. Density (cells/mm2) of T and B lymphocytes in situ decreased with age. T cells and macrophages preferentially localized near or within epithelial bilayers, rather than the intralobular stroma. M2 macrophage density was higher than M1 macrophage density and this difference was due to higher density of M2 in the intralobular stroma. Transcriptional signature analyses suggested age-dependent decline in adaptive immune cell populations and functions and increased innate immune cell activity. T cells and macrophages are so intimately associated with the epithelia that they are embedded within the bilayer, suggesting an important role for immune-epithelial cell interactions. Age-associated decreased T cell density in peri-epithelial regions, and increased M2 macrophage density in intralobular stroma suggests the emergence of a tissue microenvironment that is simultaneously immune-senescent and immunosuppressive with age.


Subject(s)
Aging/immunology , Breast/immunology , Macrophages/immunology , T-Lymphocytes/immunology , Adult , Age Factors , Aged , Breast Neoplasms/immunology , Female , Flow Cytometry , Healthy Volunteers , Humans , Immune Tolerance , Immunohistochemistry , Machine Learning , Middle Aged
4.
BMC Cancer ; 21(1): 239, 2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33676425

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) are some of the most abundant components of the tumour microenvironment. A recent study suggested that in some cancers, CAFs express programmed death ligand 1 (PD-L1), which can act as a prognostic marker. The aim of this study was to investigate the clinicopathological significance of CAF PD-L1 expression in patients with triple-negative breast cancer (TNBC) and to identify the most suitable primary antibody for immunostaining for CAF PD-L1. METHODS: Immunohistochemical staining (primary antibodies of 73-10, SP142, and E1L3N) and tissue microarrays were used to analyse the expression profiles of PD-L1 in CAF in 61 patients with TNBC who underwent surgery. Overall survival (OS) was compared based on CAF PD-L1 expression, and the risk factors for OS were analysed. The relationship between clinicopathological parameters and survival was also examined. RESULTS: Thirty-four (55.7%) patients were positive for CAF PD-L1 (73-10) expression. Compared with CAF PD-L1 negativity, there was a significant correlation between CAF PD-L1 positivity and better OS (p = 0.029). CAF PD-L1 expression, evaluated using SP-142 or E1L3N, did not correlate with OS. CAF PD-L1-positivity (73-10) correlated significantly with better prognosis in multivariate analyses (hazard ratio: 0.198; 95% confidence interval: 0.044-0.891; p = 0.035). CONCLUSIONS: CAF PD-L1 expression is a novel marker for a better prognosis of patients with TNBC, and the 73-10 assay may be suitable for immunostaining CAF PD-L1.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Neoplasm Recurrence, Local/epidemiology , Triple Negative Breast Neoplasms/mortality , Tumor Microenvironment/immunology , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/analysis , B7-H1 Antigen/metabolism , Breast/immunology , Breast/pathology , Breast/surgery , Cancer-Associated Fibroblasts/metabolism , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Mastectomy , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Prognosis , Retrospective Studies , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy
5.
BMC Cancer ; 21(1): 260, 2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33691674

ABSTRACT

BACKGROUND: Tumor microenvironment (TME) is a dynamic setting and changes in TILs and their subpopulations are potential candidates to influence the metastatic process. Aim of this pilot study is to describe the changes occurring between primary breast cancers and their paired metastases in terms of TILs composition. To assess if these changes influence the process of metastasis development, we used a control group of patients. METHODS: We retrospectively identified 18 Luminal patients, for whom primary and metastatic tissue were available (cases) and 18 paired-matched patients (controls), not relapsed after at least 9 years of follow-up, and we quantified TILs and their composition (i.e. T CD8+ and CD4+/FOXP3+). The presence of TILs was defined as ≥10%. RESULTS: Our results showed that the microenvironment composition of relapsed patients was poor of TILs (median = 5%, I-III quartiles = 0.6-5%), CD8+ (2.5%, 0-5%) and CD4+/FOXP3 + (0%, 0-0.6%) in the primary tumor. Comparable results were observed in their related metastases (TILs 3.8%, 0.6-5%; CD8+ 0%, 0-1.3%; CD4+/FOXP3+ 0%,0-1.9%). On the contrary, the microenvironment in the control group was richer of TILs (5%, 5-17.5%) in comparison to cases, both in primary tumor (p = 0.035) and related metastases (p = 0.018). Although CD8+ in controls were similar to cases at primary tumor (p = 0.6498), but not at metastasis (p = 0.0223), they expressed only one part on the TILs subpopulations (p = 0.0060), while TILs in the cases at primary tumor were almost completely CD8+ (p = 0.5034). CONCLUSIONS: These findings suggest that the lack of activation of immune system in the primary tumor might influence the multifactor process of cancer progression.


Subject(s)
Breast Neoplasms/immunology , Breast/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Recurrence, Local/immunology , Tumor Microenvironment/immunology , Aged , Aged, 80 and over , Biopsy , Breast/immunology , Breast/surgery , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Case-Control Studies , Chemotherapy, Adjuvant , Disease Progression , Female , Follow-Up Studies , Humans , Mastectomy , Middle Aged , Neoplasm Recurrence, Local/pathology , Pilot Projects , Prognosis , Receptor, ErbB-2/analysis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/analysis , Receptors, Estrogen/metabolism , Retrospective Studies
6.
BMC Cancer ; 21(1): 286, 2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33726701

ABSTRACT

BACKGROUND: In this study, we investigated CD20+ TILs in triple-negative breast cancer (TNBC) and their relationship with T lymphocyte subsets (CD4+, CD8+, CD25+, and FOXP3+), including their combined prognostic value using an immunohistochemical staining method. METHODS: We investigated 107 patients with TNBC for whom a full-face section stained by hematoxylin and eosin between 2006 and 2018 at Dokkyo Medical University Hospital was available. RESULTS: The strongest association of infiltrating CD20+ TILs was with CD4+ TILs. There was a significant relationship between CD20+ and CD4+ TILs (r = 0.177; p < 0.001), CD8+ TILs (r = 0.085; p = 0.002), and FOXP3+ TILs (r = 0.0043; p = 0.032). No significant relationships were observed between the CD20+ and CD25+ TILs (r = 0.012; p = 0.264). Multivariate analysis revealed that only the CD20+/FOXP3 ratio was an independent factor for relapse-free survival (p < 0.001) and overall survival (p < 0.001). Patients with tumors highly infiltrated by CD4+, CD8+, and CD20+ TILs had a good prognosis. In contrast, those with tumors weakly infiltrated by CD20+ TILs but highly infiltrated by CD25+ and FOXP3+ TILs had a poor prognosis. CONCLUSIONS: CD20+ TILs may support an increase in CD4+ and CD8+ TILs, which altered the anti-tumor response, resulting in a positive prognosis. CD20+ TILs correlated with FOXP3+ Treg lymphocytes, which were reported to be correlated with a poor prognosis. Our study suggested that TIL-B cells have dual and conflicting roles in TIL-T immune reactions in TNBC.


Subject(s)
Carcinoma/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Recurrence, Local/epidemiology , Triple Negative Breast Neoplasms/therapy , B-Lymphocytes/immunology , Breast/cytology , Breast/immunology , Breast/pathology , Carcinoma/immunology , Carcinoma/mortality , Carcinoma/pathology , Chemotherapy, Adjuvant/methods , Disease-Free Survival , Female , Humans , Immunohistochemistry , Mastectomy , Middle Aged , Neoadjuvant Therapy/methods , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Prognosis , Risk Assessment/methods , T-Lymphocyte Subsets/immunology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
7.
BMC Cancer ; 21(1): 645, 2021 May 31.
Article in English | MEDLINE | ID: mdl-34059009

ABSTRACT

BACKGROUND: Breast cancer (BRCA) is a malignant tumor with high morbidity and mortality, which is a threat to women's health worldwide. Ferroptosis is closely related to the occurrence and development of breast cancer. Here, we aimed to establish a ferroptosis-related prognostic gene signature for predicting patients' survival. METHODS: Gene expression profile and corresponding clinical information of patients from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database. The Least absolute shrinkage and selection operator (LASSO)-penalized Cox regression analysis model was utilized to construct a multigene signature. The Kaplan-Meier (K-M) and Receiver Operating Characteristic (ROC) curves were plotted to validate the predictive effect of the prognostic signature. Gene Ontology (GO) and Kyoto Encyclopedia of Genes, Genomes (KEGG) pathway and single-sample gene set enrichment analysis (ssGSEA) were performed for patients between the high-risk and low-risk groups divided by the median value of risk score. RESULTS: We constructed a prognostic signature consisted of nine ferroptosis-related genes (ALOX15, CISD1, CS, GCLC, GPX4, SLC7A11, EMC2, G6PD and ACSF2). The Kaplan-Meier curves validated the fine predictive accuracy of the prognostic signature (p < 0.001). The area under the curve (AUC) of the ROC curves manifested that the ferroptosis-related signature had moderate predictive power. GO and KEGG functional analysis revealed that immune-related responses were largely enriched, and immune cells, including activated dendritic cells (aDCs), dendritic cells (DCs), T-helper 1 (Th1), were higher in high-risk groups (p < 0.001). Oppositely, type I IFN response and type II IFN response were lower in high-risk groups (p < 0.001). CONCLUSION: Our study indicated that the ferroptosis-related prognostic signature gene could serve as a novel biomarker for predicting breast cancer patients' prognosis. Furthermore, we found that immunotherapy might play a vital role in therapeutic schedule based on the level and difference of immune-related cells and pathways in different risk groups for breast cancer patients.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/genetics , Breast Neoplasms/mortality , Ferroptosis/genetics , Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Breast/immunology , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Datasets as Topic , Female , Ferroptosis/drug effects , Ferroptosis/immunology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Kaplan-Meier Estimate , Middle Aged , Predictive Value of Tests , Prognosis , Protein Interaction Maps/drug effects , Protein Interaction Maps/genetics , ROC Curve , Risk Assessment/methods
8.
Future Oncol ; 17(8): 955-963, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33538176

ABSTRACT

Metaplastic breast cancer (MPBC) is a rare and aggressive tumor type in great need of satisfactory therapies. Although most cases of MPBC are 'triple negative', they are nonetheless related to worse outcomes compared with other triple-negative invasive tumors. MPBC presents high levels of genetic and molecular heterogeneity, suggesting that novel targeted therapies can be exploited. Overexpression of PD-L1 and high levels of tumor-infiltrating lymphocytes have also been observed in these tumors, suggesting a role for immunotherapy. We present an updated literature revision on clinical, histopathological and molecular features of MPBC and their significance to prognosis and therapy options. We discuss emerging efforts to improve and personalize prognostic and therapeutic approaches, exploiting the molecular signature of MPBC with targeted therapies and immunotherapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/therapy , Genetic Heterogeneity , Mastectomy/statistics & numerical data , Neoplasm Recurrence, Local/epidemiology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast/immunology , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Chemotherapy, Adjuvant/methods , Chemotherapy, Adjuvant/statistics & numerical data , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Neoadjuvant Therapy/methods , Neoadjuvant Therapy/statistics & numerical data , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/prevention & control , Prognosis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
9.
J Mammary Gland Biol Neoplasia ; 25(4): 417-432, 2020 12.
Article in English | MEDLINE | ID: mdl-33590360

ABSTRACT

Multiplex immunofluorescence (mIF) allows simultaneous antibody-based detection of multiple markers with a nuclear counterstain on a single tissue section. Recent studies have demonstrated that mIF is becoming an important tool for immune profiling the tumor microenvironment, further advancing our understanding of the interplay between cancer and the immune system, and identifying predictive biomarkers of response to immunotherapy. Expediting mIF discoveries is leading to improved diagnostic panels, whereas it is important that mIF protocols be standardized to facilitate their transition into clinical use. Manual processing of sections for mIF is time consuming and a potential source of variability across numerous samples. To increase reproducibility and throughput we demonstrate the use of an automated slide stainer for mIF incorporating tyramide signal amplification (TSA). We describe two panels aimed at characterizing the tumor immune microenvironment. Panel 1 included CD3, CD20, CD117, FOXP3, Ki67, pancytokeratins (CK), and DAPI, and Panel 2 included CD3, CD8, CD68, PD-1, PD-L1, CK, and DAPI. Primary antibodies were first tested by standard immunohistochemistry and single-plex IF, then multiplex panels were developed and images were obtained using a Vectra 3.0 multispectral imaging system. Various methods for image analysis (identifying cell types, determining cell densities, characterizing cell-cell associations) are outlined. These mIF protocols will be invaluable tools for immune profiling the tumor microenvironment.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/immunology , Fluoroimmunoassay/methods , Image Processing, Computer-Assisted/methods , Tumor Microenvironment/immunology , Biomarkers, Tumor/metabolism , Breast/immunology , Breast/pathology , Breast Neoplasms/pathology , Female , Fluorescent Dyes/chemistry , Fluoroimmunoassay/instrumentation , Humans , Reproducibility of Results , Tissue Array Analysis/instrumentation , Tissue Array Analysis/methods
10.
Breast Cancer Res ; 22(1): 104, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004039

ABSTRACT

BACKGROUND: Systemic inflammation may play a role in shaping breast composition, one of the strongest risk factors for breast cancer. Pubertal development presents a critical window of breast tissue susceptibility to exogenous and endogenous factors, including pro-inflammatory markers. However, little is known about the role of systemic inflammation on adolescent breast composition and pubertal development among girls. METHODS: We investigated associations between circulating levels of inflammatory markers (e.g., interleukin-6 (IL-6), tumor necrosis factor receptor 2 (TNFR2), and C-reactive protein (CRP)) at Tanner stages 2 and 4 and breast composition at Tanner stage 4 in a cohort of 397 adolescent girls in Santiago, Chile (Growth and Obesity Cohort Study, 2006-2018). Multivariable linear models were used to examine the association between breast composition and each inflammatory marker, stratifying by Tanner stage at inflammatory marker measurement. Accelerated failure time models were used to evaluate the association between inflammatory markers concentrations at each Tanner stage and time to menarche. RESULTS: In age-adjusted linear regression models, a doubling of TNFR2 at Tanner 2 was associated with a 26% (95% CI 7-48%) increase in total breast volume at Tanner 4 and a 22% (95% CI 10-32%) decrease of fibroglandular volume at Tanner 4. In multivariable models further adjusted for body fatness and other covariates, these associations were attenuated to the null. The time to menarche was 3% (95% CI 1-5%) shorter among those in the highest quartile of IL-6 at Tanner 2 relative to those in the lowest quartile in fully adjusted models. Compared to those in the lowest quartile of CRP at Tanner 4, those in the highest quartile experienced 2% (95% CI 0-3%) longer time to menarche in multivariable models. CONCLUSIONS: Systemic inflammation during puberty was not associated with breast volume or breast density at the conclusion of breast development among pubertal girls after adjusting for body fatness; however, these circulating inflammation biomarkers, specifically CRP and IL-6, may affect the timing of menarche onset.


Subject(s)
Biomarkers/blood , Breast Density , Breast/immunology , Inflammation/immunology , Menarche , Puberty , Breast/growth & development , Breast/metabolism , C-Reactive Protein/metabolism , Child , Cohort Studies , Female , Humans , Inflammation/metabolism , Interleukin-6/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Risk Factors , Sexual Maturation
11.
Breast Cancer Res ; 22(1): 140, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33357231

ABSTRACT

BACKGROUND: Tumour-infiltrating lymphocytes (TILs) are of important prognostic and predictive value in human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC) and triple-negative breast cancer (TNBC), but their clinical relevance in oestrogen receptor-positive/HER2-negative (ER+/HER2-) remains unknown. The primary study aim was to analyse the prognostic effect of TILs on the BC-free interval (BCFi) in premenopausal patients stratified by BC subtypes. The secondary aim was to investigate if TILs are predictive of tamoxifen (TAM) benefit. METHODS: Archival tissues from primary breast tumours were collected from patients from the SBII:2pre trial, in which 564 premenopausal women were randomised to 2 years of adjuvant TAM or no systemic treatment, regardless of hormone receptor status. TILs were scored on whole tissue sections from 447 patients with available ER status. Tumours were divided into ER+/HER2-, HER2+ and TNBC subtypes by immunohistochemistry and in situ hybridisation. The prognostic value of TILs was analysed in systemically untreated patients (n = 221); the predictive information was investigated in the ER+ subgroup (n = 321) by cumulative incidence curves and Cox regression analyses. The median follow-up was 28 years. RESULTS: High (≥ 50%) infiltration of TILs was a favourable prognostic factor in terms of BCFi (univariable analysis: hazard ratioBCFi (HRBCFi) 0.40; 95% confidence interval (CI) 0.22-0.71; P = 0.002). Similar effects were observed across all BC subtypes. The effect of adjuvant TAM was stronger in patients with ER+ tumours and TILs < 50% (HRBCFi 0.63; 95% CI 0.47-0.84; P = 0.002) than in patients with high immune infiltration (≥ 50%) (HRBCFi 0.84; 95% CI (0.24-2.86); P = 0.77). However, evidence for differential effects of TAM in categories of TILs, i.e. interaction, was weak. CONCLUSIONS: We demonstrate a long-term favourable prognostic value of high infiltration of TILs in a cohort of premenopausal BC patients and the positive prognostic effect was extended to the ER+/HER2- subgroup. A beneficial effect of TAM in ER+ patients was observed in patients with tumours of low TIL infiltration, but evidence for a treatment predictive effect was weak. TRIAL REGISTRATION: This trial is registered in the ISRCTN database, trial ID: ISRCTN12474687 .


Subject(s)
Breast Neoplasms/mortality , Breast/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Recurrence, Local/epidemiology , Tamoxifen/pharmacology , Adult , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Breast/immunology , Breast/surgery , Breast Neoplasms/diagnosis , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Chemotherapy, Adjuvant/methods , Disease-Free Survival , Drug Resistance, Neoplasm/immunology , Female , Follow-Up Studies , Humans , Immunohistochemistry , Mastectomy , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Neoplasm Staging , Premenopause , Prognosis , Prospective Studies , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Receptors, Estrogen/metabolism , Retrospective Studies , Tamoxifen/therapeutic use
12.
Breast Cancer Res ; 22(1): 134, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33267869

ABSTRACT

BACKGROUND: Inflammatory breast cancer (IBC) is a rare but aggressive carcinoma characterized by severe erythema and edema of the breast, with many patients presenting in advanced metastatic disease. The "inflammatory" nature is not due to classic immune-mediated inflammation, but instead results from tumor-mediated blockage of dermal lymphatic ducts. Previous work has shown that expression of PD-L1 on tumor cells can suppress T cell activation in triple-negative (TN) non-IBC breast cancer. In the present work, we investigated immune parameters in peripheral blood of metastatic IBC patients to determine whether cellular components of the immune system are altered, thereby contributing to pathogenesis of the disease. These immune parameters were also compared to PD-1 and PD-L1 expression in IBC tumor biopsies. METHODS: Flow cytometry-based immune phenotyping was performed using fresh peripheral blood from 14 stage IV IBC patients and compared to 11 healthy age-similar control women. Immunohistochemistry for CD20, CD3, PD-1, and PD-L1 was performed on tumor biopsies of these metastatic IBC patients. RESULTS: IBC patients with Stage IV disease had lymphopenia with significant reductions in circulating T, B, and NK cells. Reductions were observed in all subsets of CD4+ T cells, whereas reductions in CD8+ T cells were more concentrated in memory subsets. Immature cytokine-producing CD56bright NK cells expressed higher levels of FcγRIIIa and cytolytic granule components, suggesting accelerated maturation to cytolytic CD56dim cells. Immunohistochemical analysis of tumor biopsies demonstrated moderate to high expression of PD-1 in 18.2% of patients and of PD-L1 in 36.4% of patients. Interestingly, a positive correlation was observed between co-expression levels of PD-L1 and PD-1 in tumor biopsies, and higher expression of PD-L1 in tumor biopsies correlated with higher expression of cytolytic granule components in blood CD4+ T cells and CD56dim NK cells, and higher numbers of CD8+ effector memory T cells in peripheral blood. PD-1 expression in tumor also correlated with increased infiltration of CD20+ B cells in the tumor. CONCLUSIONS: Our results suggest that while lymphocyte populations are severely compromised in stage IV IBC patients, an immune response toward the tumor had occurred in some patients, providing biological rationale to evaluate PD-1/PD-L1 immunotherapies for IBC.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma/immunology , Inflammatory Breast Neoplasms/immunology , T-Lymphocytes/immunology , Adult , Aged , Antigens, CD20/analysis , Antigens, CD20/metabolism , B7-H1 Antigen/analysis , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Biopsy , Breast/immunology , Breast/pathology , CD3 Complex/analysis , CD3 Complex/metabolism , Carcinoma/blood , Carcinoma/diagnosis , Carcinoma/secondary , Case-Control Studies , Female , Flow Cytometry , Humans , Immunity, Cellular , Immunohistochemistry , Immunophenotyping/methods , Inflammatory Breast Neoplasms/blood , Inflammatory Breast Neoplasms/diagnosis , Inflammatory Breast Neoplasms/pathology , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Middle Aged , Neoplasm Staging , Programmed Cell Death 1 Receptor/analysis , Programmed Cell Death 1 Receptor/metabolism , Retrospective Studies , T-Lymphocytes/metabolism
13.
Cancer Immunol Immunother ; 69(9): 1841-1854, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32350594

ABSTRACT

OBJECTIVE: Mesenchymal stem cells (MSCs), one of the most important stromal cells in the tumor microenvironment, play a major role in the immunomodulation and development of tumors. In contrast to immunomodulatory effects of bone marrow-derived MSCs, resident MSCs were not well studied in tumor. The aim of this study was to compare the immunomodulatory properties and protein secretion profiles of MSCs isolated from breast tumor (T-MSC) and normal breast adipose tissue (N-MSC). MATERIALS AND METHODS: T-MSCs and N-MSCs were isolated by the explant culture method and characterized, and their immunomodulatory function was assessed on peripheral blood lymphocytes (PBLs) by evaluating the effects of MSC conditioned media on the proliferation and induction of some cytokines and regulatory T cells (Tregs) by BrdU assay, ELISA, and flow cytometry. In addition, we compared the secretion of indoleamine 2,3-dioxygenase (IDO), vascular endothelial growth factor (VEGF), matrix metallopeptidase (MMP)-2, MMP-9, and Galectin-1. RESULTS: T-MSCs showed a higher secretion of transforming growth factor beta (TGF-ß), prostaglandin E2 (PGE2), IDO, and VEGF and lower secretion of MMP-2 and MMP-9 compared with N-MSCs. However, no significant difference was found in the secretion of interferon gamma (IFN-γ), interleukin 10 (IL10), IL4, IL17, and Galectin-1 in T-MSCs and N-MSCs. The immunomodulatory effect of soluble factors on PBLs showed that T-MSCs, in contrast to N-MSCs, stimulate PBL proliferation. Importantly, the ability of T-MSCs to induce IL10, TGF-ß, IFN-γ, and PGE2 was higher than that of N-MSCs. In addition, T-MSCs and N-MSCs exhibited no significant difference in Treg induction. CONCLUSION: MSCs educated in stage II breast cancer and normal breast adipose tissue, although sharing a similar morphology and immunophenotype, exhibited a clearly different profile in some immunomodulatory functions and protein secretions.


Subject(s)
Adipose Tissue/immunology , Breast Neoplasms/immunology , Breast/immunology , Immunomodulation/immunology , Mesenchymal Stem Cells/immunology , Adult , Case-Control Studies , Cell Proliferation/physiology , Cytokines/immunology , Dinoprostone/immunology , Female , Humans , T-Lymphocytes, Regulatory/immunology , Vascular Endothelial Growth Factor A/immunology
14.
Cancer Immunol Immunother ; 69(5): 799-811, 2020 May.
Article in English | MEDLINE | ID: mdl-32055918

ABSTRACT

The underlying basis for cancer immune evasion is important for effective immunotherapy and prognosis in breast cancers. Human leucocyte antigens (HLA)-I comprising three classical antigens (HLA-A, -B and -C) is mandatory for anti-tumor immunity. Its loss occurred frequently in many cancers resulting in effective immune evasion. Most studies examined HLA-I as a whole. Alterations in specific locus could have different clinical ramifications. Hence, we evaluated the expression of the three HLA-I loci in a large cohort of breast cancers. Low expression of HLA-A, -B and -C were found in 71.1%, 66.3%, and 60.2% of the cases. Low and high expression in all loci was found in 48.3% and 17.9% of the cases respectively. The remaining showed high expression in one or two loci. Cases with all HLA high expression (all HLA high) was frequent in the ER-HER2- (27.4%) and ER-HER2+ (23.1%) cases and was associated with characteristic pathologic features related to these tumor (higher grade, necrosis, high tumor infiltrating lymphocyte (TIL), pT stage, low hormonal receptor, high basal marker expression) (p ≤ 0.019). Interestingly, in HER2+ cancers, only cases with all HLA high and high TIL showed significantly better survival. In node positive cancers, concordant high HLA expression in primary tumors and nodal metastases was favorable prognostically (DFS: HR = 0.741, p < 0.001; BCSS: HR = 0.699, p = 0.003). The data suggested an important clinical value of a combined analysis on the co-expression HLA-I status in both primary and metastatic tumors. This could be a potential additional key component to be incorporated into TIL evaluation for improved prognostication.


Subject(s)
Breast Neoplasms/pathology , Histocompatibility Antigens Class I/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Receptor, ErbB-2/metabolism , Adult , Aged , Aged, 80 and over , Breast/immunology , Breast/pathology , Breast/surgery , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Disease-Free Survival , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic/immunology , Genes, MHC Class I/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Kaplan-Meier Estimate , Mastectomy , Middle Aged , Prognosis , Tissue Array Analysis , Young Adult
15.
Immunol Cell Biol ; 98(10): 883-896, 2020 11.
Article in English | MEDLINE | ID: mdl-32713010

ABSTRACT

Ex vivo mammary explant systems are an excellent model to study interactions between epithelium and stromal cell types because they contain physiologically relevant heterotypic interactions in the background of genetically diverse patients. The intact human mammary tissue, termed patient-derived explant (PDE), can be used to investigate cellular responses to a wide variety of external stimuli in situ. For this study, we examined the impact of cytokines or environmental chemicals on macrophage phenotypes. We demonstrate that we can polarize macrophages within human breast tissue PDEs toward M1 or M2 through the addition of interferon-γ (IFNγ) + lipopolysaccharide (LPS) or interleukin (IL)-4 + IL-13, respectively. Elevated expression levels of M(IFNγ + LPS) markers (HLADRA and CXCL10) or M(IL-4 + IL-13) markers (CD209 and CCL18) were observed in cytokine-treated tissues. We also examined the impact of the endocrine-disrupting chemical, benzophenone-3, on PDEs and measured significant, yet varying effects on macrophage polarization. Furthermore, a subset of the PDEs respond to IL-4 + IL-13 through downregulation of E-cadherin and upregulation of vimentin which is reminiscent of epithelial-to-mesenchymal transition (EMT) changes. Finally, we were able to show immortalized nonmalignant breast epithelial cells can exhibit EMT characteristics when exposed to growth factors secreted by M(IL-4 + IL-13) macrophages. Taken together, the PDE model system is an outstanding preclinical model to study early tissue-resident immune responses and effects on epithelial and stromal responses to stimuli found both endogenously in the breast and exogenously as a result of exposures.


Subject(s)
Breast/immunology , Environmental Exposure , Macrophage Activation , Benzophenones/adverse effects , Breast/drug effects , Cell Polarity , Endocrine Disruptors/adverse effects , Female , Humans , Macrophages/cytology , Tissue Culture Techniques
16.
Mod Pathol ; 33(9): 1773-1782, 2020 09.
Article in English | MEDLINE | ID: mdl-32341499

ABSTRACT

Ductal carcinoma in situ (DCIS) associated stromal changes and influx of immune cells might be mediators of progression to invasive breast cancer. We studied the interaction between DCIS-associated stromal changes, and immune cell distribution and composition in a well-characterized patient cohort. We included 472 patients with DCIS. The presence of stromal changes, signs of regression, and DCIS-associated immune cell position were determined on hematoxylin and eosin-stained slides. Immune cell composition was characterized by immunohistochemistry (CD4, CD8, CD20, CD68, and FOXP3). The number of intraductal immune cells was quantified per mm2. The interaction between stromal changes, signs of DCIS regression, immune cell composition and location was explored. Stromal changes and signs of DCIS regression were identified in 30 and 7% of the patients, respectively. Intraductal immune cells mainly comprised CD68+ macrophages and CD8+ T cells. Patients with stromal changes had significantly less influx of immune cells within the duct. DCIS regression was associated with an increased number of intraductal FOXP3+ T cells. The highest number of intraductal CD8+ T cells was seen in the ER+ HER2+ subtype. We suggest that DCIS-associated stromal changes prevent the interaction between immune cells and DCIS cells. However, in case of DCIS regression, we surmise a direct interaction between DCIS cells and immune cells, in particular FOXP3+ cells. Furthermore, the increased number of intraductal CD8+ T cells in the ER+ HER2+ DCIS subtype suggests a subtype-specific immune response, which is likely to play a role in the distinct biological behavior of different DCIS subtypes.


Subject(s)
Breast Neoplasms/pathology , Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Stromal Cells/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Breast/immunology , Breast/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/immunology , Carcinoma, Intraductal, Noninfiltrating/metabolism , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Stromal Cells/immunology , Tumor Microenvironment/immunology
17.
BMC Cancer ; 20(1): 598, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32590956

ABSTRACT

BACKGROUND: Lymph node metastasis is more likely in early-stage breast cancer with lower tumor-infiltrating lymphocyte (TIL) density. Therefore, we investigated the correlation between TILs and lymph node metastasis in cT1 breast cancer patients undergoing surgery and the usefulness of TILs in predicting sentinel lymph node metastasis (SLNM) in cT1N0M0 breast cancer. METHODS: We investigated 332 breast cancer patients who underwent surgery as the first-line treatment after preoperative diagnosis of cT1. A positive diagnosis of SLNM as an indication for axillary clearance was defined as macrometastasis in the sentinel lymph node (SLN) (macrometastasis: tumor diameter > 2 mm). Semi-quantitative evaluation of lymphocytes infiltrating the peritumoral stroma as TILs in primary tumor biopsy specimens prior to treatment was conducted. RESULTS: For SLN biopsy (SLNB), a median of 2 (range, 1-8) SLNs were pathologically evaluated. Sixty cases (19.4%) of SLNM (macrometastasis: 46, micrometastasis: 16) were observed. Metastasis was significantly greater in breast cancers with tumor diameter > 10 mm than in those with diameter ≤ 10 mm (p = 0.016). Metastasis was significantly associated with lymphatic invasion (p < 0.001). These two clinicopathological factors correlated with SLNM even in patients diagnosed with cN0 (tumor size; p = 0.017, lymphatic invasion; p = 0.002). Multivariate analysis for SLNM predictors revealed lymphatic invasion (p = 0.008, odds ratio [OR] = 2.522) and TILs (p < 0.001, OR = 0.137) as independent factors. CONCLUSIONS: Our results suggest a correlation between lymph node metastasis and tumor immune-microenvironment in cT1 breast cancer. TIL density may be a predictor of SLNM in breast cancer without lymph node metastasis on preoperative imaging.


Subject(s)
Breast Neoplasms/pathology , Breast/pathology , Lymphatic Metastasis/diagnosis , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Aged , Breast/cytology , Breast/immunology , Breast/surgery , Breast Neoplasms/immunology , Breast Neoplasms/surgery , Female , Humans , Lymphatic Metastasis/immunology , Mastectomy , Middle Aged , Neoplasm Staging , Preoperative Period , Prognosis , Risk Factors , Sentinel Lymph Node/diagnostic imaging , Sentinel Lymph Node/pathology , Sentinel Lymph Node Biopsy , Tomography, X-Ray Computed , Tumor Microenvironment/immunology
18.
BMC Cancer ; 20(1): 766, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32799825

ABSTRACT

BACKGROUND: Sphingosine-1-phosphate receptor (S1PR1) is involved in vascular development, a key process in tumorigenesis. This study aimed to evaluate its roles in tumor development and prognosis. METHODS: S1PR1 expression levels were analyzed using TIMER and Oncomine database, and the prognostic significance of S1PR1 was assessed using PrognoScan and Kaplan-Meier plotter databases. The relationship between S1PR1 and tumor-infiltrated immune cells was analyzed using TIMER. RESULTS: S1PR1 expression was remarkably lower in breast and lung cancer tissues than in the corresponding normal tissues. Lower expression was related to poor overall survival and disease-free survival in breast invasive carcinoma (BRCA), lung adenocarcinoma (LUAD), and lung squamous cell carcinoma (LUSC). A functional network analysis confirmed the function of S1PR1 in regulating vasculogenesis. In addition, S1PR1 levels were significantly negative with regard to the tumor purity of BRCA (r = - 0.508, P = 1.76e-66), LUAD (r = - 0.353, P = 6.05e-16), and LUSC (r = - 0.402, P = - 5.20e-20). Furthermore, S1PR1 levels were significantly positive with regard to infiltrating CD8+ (r = 0.38, P = 5.91e-35) and CD4+ T cells (r = 0.335, P = 1.03e-26), macrophages (r = 0.219, P = 3.67e-12), neutrophils (r = 0.168, P = 2.03e-7), and dendritic cells (DCs) (r = 0.208, P = 9.14e-11) in BRCA; S1PR1 levels were significantly positive with regard to CD8+ T cells (r = 0.308, P = 3.61e-12), macrophages (r = 0.376, P = 1.01e-17), neutrophils (r = 0.246, P = 4.15e-8), and DCs (r = 0.207, P = 4.16e-6) in LUAD; and positive with regard to B cells (r = 0.356, P = 1.57e-15), CD8+ (r = 0.459, P = 3.83e-26) and CD4+ T cells (r = 0.338, P = 3.98e-14), macrophages (r = 0.566, P = 2.61e-45), neutrophils (r = 0.453, P = 1.79e-25), and DCs (r = 0.56, P = 2.12e-40) in LUSC. CONCLUSIONS: S1PR1 levels are positively correlated with multiple immune markers in breast and lung cancer. These observed correlations between S1PR1 and the prognosis and immune cell infiltration provide a foundation for further research on its immunomodulatory role in cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Sphingosine-1-Phosphate Receptors/metabolism , Biomarkers, Tumor/analysis , Breast/cytology , Breast/immunology , Breast/pathology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Carcinogenesis/immunology , Carcinogenesis/pathology , Datasets as Topic , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Lung/cytology , Lung/immunology , Lung/pathology , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Male , Prognosis , Sphingosine-1-Phosphate Receptors/analysis , Tumor Microenvironment/immunology
19.
J Surg Res ; 256: 645-656, 2020 12.
Article in English | MEDLINE | ID: mdl-32810665

ABSTRACT

BACKGROUND: Although previous experiments have implicated sphingosine-1-phosphate (S1P) as a links between immune reactions and cancer progression, the exact mechanism of this interaction has not comprehensively studied in clinical human samples. This study sought to evaluate the S1P regulation by sphingosine kinase 1 (SPHK1), an S1P-producing enzyme, in the immunity/immuno-reactivity of clinical human breast cancer surgical specimens. METHODS: S1P levels were examined in tumor, peritumoral, and normal human breast samples using mass spectrometry. Genomics Data Commons data portal of The Cancer Genome Atlas cohort was used to assess the expression of S1P-related and immune-related genes. RESULTS: S1P levels were significantly higher in tumor samples compared to peritumoral (P < 0.05) or normal human breast samples (P < 0.001). SPHK1 gene expression was elevated in tumoral samples compared to normal breast samples (P < 0.01). Furthermore, the elevated expression of SPHK1 in breast cancer tissue was associated with an increased expression of the different kinds of immune-related genes, such as CD68, CD163, CD4, and FOXP3 (forkhead box P3), in HER2-negative breast cancer. Network analysis showed the central role of SPHK1 in the interaction of S1P signaling and expression of immune cell-related proteins. CONCLUSIONS: We demonstrated that S1P is mainly produced by tumor tissue, rather than peritumoral tissue, in breast cancer patients. Our data revealed the involvement of S1P signaling in the regulation of immune-related genes, suggesting the links between S1P and complicated immune-cancer interactions in breast cancer patients.


Subject(s)
Breast Neoplasms/immunology , Breast/pathology , Gene Expression Regulation, Neoplastic/immunology , Lysophospholipids/analysis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/analogs & derivatives , Breast/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cohort Studies , Datasets as Topic , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/analysis , Protein Interaction Maps/genetics , Protein Interaction Maps/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Spectrometry, Mass, Electrospray Ionization , Sphingosine/analysis , Sphingosine/metabolism , Tandem Mass Spectrometry
20.
Acta Oncol ; 59(9): 1103-1109, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32459128

ABSTRACT

BACKGROUND: As tumors maintain an inflammatory microenvironment, anti-inflammatory medication can be useful in cancer therapy. We have previously shown an association with improved survival in melanoma for use of the H1-antihistamines desloratadine and loratadine, and here we examine use of H1-antihistamines and breast cancer mortality. MATERIAL AND METHODS: We investigated use of the six major H1-antihistamines (cetirizine, clemastine, desloratadine, ebastine, fexofenadine and loratadine) and breast cancer-specific and overall mortality in a nation-wide register-based study of all 61,627 Swedish women diagnosed with breast cancer 2006-2013. Both peri- and post-diagnostic antihistamine use was analyzed using Cox regression models. Analyses were stratified for age and subgroup analyses based on estrogen receptor status and menopausal status were performed. RESULTS: We found a consistently improved survival of desloratadine users (HR = 0.67; 95% CI 0.55-0.81, p < .001), as well as of loratadine users (HR = 0.80; 95% CI 0.67-0.95, p = .012), relative to nonusers, regardless of patient age, menopause, estrogen receptor status or stage of the tumor, or whether breast cancer-specific or overall survival was analyzed. The survival of users of other antihistamines varied relative to non-users. CONCLUSION: Based on their safety and current use within the patient population, together with our observations, we suggest the initiation of trials of desloratadine and loratadine as treatment of breast cancer as well as studies of the mechanism behind their possible effect. Further studies on any effects of other H1-antihistamines may also be merited, as well as of H1-antihistamine use and survival in other malignancies.


Subject(s)
Breast Neoplasms/mortality , Drug Prescriptions/statistics & numerical data , Histamine H1 Antagonists, Non-Sedating/administration & dosage , Loratadine/analogs & derivatives , Breast/drug effects , Breast/immunology , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Female , Follow-Up Studies , Humans , Loratadine/administration & dosage , Middle Aged , Receptors, Estrogen/metabolism , Registries/statistics & numerical data , Risk Assessment/statistics & numerical data , Risk Factors , Survival Analysis , Survival Rate , Sweden/epidemiology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL