Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
BMC Cancer ; 18(1): 123, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29390966

ABSTRACT

BACKGROUND: The tumour microenvironment consists of malignant cells, stroma and immune cells. In women with large and locally advanced breast cancers (LLABCs) undergoing neoadjuvant chemotherapy (NAC), tumour-infiltrating lymphocytes (TILs), various subsets (effector, regulatory) and cytokines in the primary tumour play a key role in the induction of tumour cell death and a pathological complete response (pCR) with NAC. Their contribution to a pCR in nodal metastases, however, is poorly studied and was investigated. METHODS: Axillary lymph nodes (ALNs) (24 with and 9 without metastases) from women with LLABCs undergoing NAC were immunohistochemically assessed for TILs, T effector and regulatory cell subsets, NK cells and cytokine expression using labelled antibodies, employing established semi-quantitative methods. IBM SPSS statistical package (21v) was used. Non-parametric (paired and unpaired) statistical analyses were performed. Univariate and multivariate regression analyses were carried out to establish the prediction of a pCR and Spearman's Correlation Coefficient was used to determine the correlation of immune cell infiltrates in ALN metastatic and primary breast tumours. RESULTS: In ALN metastases high levels of TILs, CD4+ and CD8+ T and CD56+ NK cells were significantly associated with pCRs.. Significantly higher levels of Tregs (FOXP3+, CTLA-4+) and CD56+ NK cells were documented in ALN metastases than in the corresponding primary breast tumours. CD8+ T and CD56+ NK cells showed a positive correlation between metastatic and primary tumours. A high % CD8+ and low % FOXP3+ T cells and high CD8+: FOXP3+ ratio in metastatic ALNs (tumour-free para-cortex) were associated with pCRs. Metastatic ALNs expressed high IL-10, low IL-2 and IFN-ϒ. CONCLUSIONS: Our study has provided new data characterising the possible contribution of T effector and regulatory cells and NK cells and T helper1 and 2 cytokines to tumour cell death associated with NAC in ALNs. TRIAL REGISTRATION: The Trial was retrospectively registered. Study Registration Number is ISRCTN00407556 .


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Adult , Aged , Axilla/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CD56 Antigen/genetics , CTLA-4 Antigen/genetics , Cell Death/genetics , Female , Forkhead Transcription Factors/genetics , Humans , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/pathology , Middle Aged , Neoadjuvant Therapy , Neoplasm Staging , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Th1 Cells/drug effects , Th2 Cells/drug effects , Tumor Microenvironment/drug effects
2.
J Immunol Res ; 2017: 1049023, 2017.
Article in English | MEDLINE | ID: mdl-28913366

ABSTRACT

The tumour microenvironment consists of malignant cells, stroma, and immune cells. The role of adaptive immunity in inducing a pathological complete response (pCR) in breast cancer with neoadjuvant chemotherapy (NAC) is well studied. The contribution of innate immunity, however, is poorly documented. Breast tumours and axillary lymph nodes (ALNs) from 33 women with large and locally advanced breast cancers (LLABCs) undergoing NAC were immunohistochemically assessed for tumour-infiltrating macrophages (TIMs: M1 and M2), neutrophils (TINs), and dendritic cells (TIDCs) using labelled antibodies and semiquantitative methods. Patients' blood neutrophils (n = 108), DCs (mDC1 and pDC), and their costimulatory molecules (n = 30) were also studied. Pathological results were classified as pCR, good (GPR) or poor (PRR). In breast and metastatic ALNs, high levels of CD163+ TIMs were significantly associated with a pCR. In blood, high levels of neutrophils were significantly associated with pCR in metastatic ALNs, whilst the % of mDC1 and pDC and expression of HLA-DR, mDC1 CD40, and CD83 were significantly reduced. NAC significantly reduced tumour DCs but increased blood DCs. PPRs to NAC had significantly reduced HLA-DR, CD40, and CD86 expression. Our study demonstrated novel findings documenting the differential but important contributions of innate immunity to pCRs in patients with LLABCs undergoing NAC.


Subject(s)
Breast Neoplasms/immunology , Dendritic Cells/immunology , Macrophages/immunology , Neutrophils/immunology , Adult , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Differentiation , Female , Follow-Up Studies , Humans , Immunity, Innate , Lymph Nodes/pathology , Middle Aged , Neoadjuvant Therapy , Neoplasm Metastasis , Neoplasm Staging , Receptors, Cell Surface/metabolism , Survival Analysis , Tumor Microenvironment
3.
J Immunol Res ; 2016: 4757405, 2016.
Article in English | MEDLINE | ID: mdl-27777963

ABSTRACT

The tumour microenvironment consists of malignant cells, stroma, and immune cells. Prominent tumour-infiltrating lymphocytes (TILs) in breast cancer are associated with a good prognosis and are predictors of a pathological complete response (pCR) with neoadjuvant chemotherapy (NAC). The contribution of different T effector/regulatory cells and cytokines to tumour cell death with NAC requires further characterisation and was investigated in this study. Breast tumours from 33 women with large and locally advanced breast cancers undergoing NAC were immunohistochemically (intratumoural, stromal) assessed for T cell subsets and cytokine expression using labelled antibodies, employing established semiquantitative methods. Prominent levels of TILs and CD4+, CD8+, and CTLA-4+ (stromal) T cells and CD8+ : FOXP3+ ratios were associated with a significant pCR; no association was seen with FOXP3+, CTLA-4+ (intratumoural), and PD-1+ T cells. NAC significantly reduced CD4+, FOXP3+, CTLA-4+ (stromal) (concurrently blood FOXP3+, CTLA-4+ Tregs), and PD-1+ T cells; no reduction was seen with CD8+ and CTLA-4+ (intratumoural) T cells. High post-NAC tumour levels of FOXP3+ T cells, IL-10, and IL-17 were associated with a failed pCR. Our study has characterised further the contribution of T effector/regulatory cells and cytokines to tumour cell death with NAC.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/therapy , Cytokines/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoadjuvant Therapy , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Adult , Breast Neoplasms/physiopathology , CD8-Positive T-Lymphocytes/immunology , Cell Death/immunology , Chemotherapy, Adjuvant , Female , Humans , Immunohistochemistry , Interleukin-17/immunology , Lymphocyte Count , Th1 Cells/immunology , Th2 Cells/immunology
4.
J Transl Med ; 13: 180, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26040463

ABSTRACT

BACKGROUND: NK cells contribute to tumour surveillance, inhibition of growth and dissemination by cytotoxicity, secretion of cytokines and interaction with immune cells. Their precise role in human breast cancer is unclear and the effect of therapy poorly studied. The purpose of our study was to characterise NK cells in women with large (≥3 cm) and locally advanced (T3-4, N1-2, M0) breast cancers (LLABCs) undergoing neoadjuvant chemotherapy (NAC) and surgery, and to ascertain their possible contribution to a pathological complete response (pCR). METHODS: Women with LLABCs (n = 25) and healthy female donors [HFDs (n = 10)] were studied. Pathological responses in the breast were assessed using established criteria. Blood samples were collected pre and post NAC and surgery. Flow cytometry and labelled monoclonal antibodies established absolute numbers (AbNs) and percentages (%) of NK cells, and expressing granzyme B/perforin and NKG2D. In vitro NK cytotoxicity was assessed and NK cells and cytokines (IL-2, INF-γ, TGF-ß) documented in tumours using immunohistochemical techniques. Data was analysed by SPSS. RESULTS: Women with LLABCs had significantly reduced AbNs (160.00 ± 40.00 cells/µl) but not % of NK cells, compared with HFDs (NK: 266.78 ± 55.00 cells/µl; p = 0.020). NAC enhanced the AbN (p = 0.001) and % (p = 0.006) of NK cells in patients with good pathological responses. Granzyme B(+)/perforin(+) cells were significantly reduced (43.41 ± 4.00%), compared with HFDs (60.26 ± 7.00%; p = 0.003). NAC increased the % in good (p = 0.006) and poor (p = 0.005) pathological responders. Pretreatment NK cytotoxicity was significantly reduced in good (37.80 ± 8.05%) and poor (22.80 ± 7.97%) responders (p = 0.001) but remained unchanged following NAC. NK-NKG2D(+) cells were unaltered and unaffected by NAC; NKG2D expression was increased in patients with a pCR (p = 0.001). Surgery following NAC was not beneficial, except in those with a pCR. Tumour-infiltrating NK cells were infrequent but increased peritumourally (p = 0.005) showing a significant correlation (p = 0.004) between CD56(+) cells and grade of response. Tumour cytokines had no effect. CONCLUSION: Women with LLABCs have inhibited blood innate immunity, variably reversed by NAC (especially with tumour pCRs), which returned to pretreatment levels following surgery. These and in situ tumour findings suggest a role for NK cells in NAC-induced breast pCR.


Subject(s)
Breast Neoplasms/blood , Breast Neoplasms/immunology , Killer Cells, Natural/immunology , Neoadjuvant Therapy , Biopsy , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , CD56 Antigen/metabolism , Carcinoma in Situ/blood , Carcinoma in Situ/drug therapy , Carcinoma in Situ/immunology , Carcinoma in Situ/surgery , Cell Death , Cytokines/metabolism , Female , Flow Cytometry , Granzymes/metabolism , Humans , K562 Cells , Leukocyte Count , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasm Staging , Perforin/metabolism , Treatment Outcome
5.
J Transl Med ; 11: 16, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23320561

ABSTRACT

BACKGROUND: Host defences play a key role in tumour growth. Some of the benefits of chemotherapy may occur through modulation of these defences. The aim of this study was to define the status of regulatory cells in women with large and locally advanced breast cancers (LLABCs) undergoing neoadjuvant chemotherapy (NAC) and surgery. METHODS: Bloods were collected from patients (n=56) before, during and following NAC, and surgery. Controls (n=10) were healthy, age-matched females donors (HFDs). Blood mononuclear cells (BMCs) were isolated and T regulatory cells (Tregs) (n=31) determined. Absolute numbers (AbNs) of Tregs and myeloid-derived suppressor cells (MDSCs) were ascertained from whole blood (n=25). Reverse transcriptase polymerase chain reaction analysis determined Treg mRNA (n=16). In vitro production of Th1, Th2 and Th17 cytokines (n=30), was documented. Patients were classified as clinical responders by magnetic resonance mammography after two cycles of NAC and as pathological responders using established criteria, following surgery. RESULTS: Patients with LLABCs had significantly increased circulating Tregs (≥ 6 fold AbN and percentage (%)) and MDSCs (≥ 1.5 fold AbN (p=0.025)). Percentage of FOXP3+ Tregs in blood predicted the response of the LLABCs to subsequent NAC (p=0.04). Post NAC blood Tregs (%) were significantly reduced in patients where tumours showed a good pathological response to NAC (p=0.05). Blood MDSCs (granulocytic, monocytic) were significantly reduced in all patients, irrespective of the pathological tumour response to chemotherapy. NAC followed by surgery failed to restore blood Tregs to normal levels. MDSCs, however, were reduced to or below normal levels by NAC alone. Invitro Th1 profile (IL-1ß, IL-2, INF-γ, TNF-α) was significantly reduced (p ≤ 0.009), whilst Th2 (IL-4, IL-5) was significantly enhanced (P ≤ 0.004). Th1 and Th2 (IL-5) were unaffected by NAC and surgery. IL-17A was significantly increased (p ≤ 0.023) but unaffected by chemotherapy and surgery. CONCLUSION: Women with LLABCs have abnormal blood regulatory cell levels (Tregs and MDSCs) and cytokine profiles (Th1, Th2, Th17). NAC followed by surgery failed to abolish the abnormal Treg and Th profiles. There was a significant correlation between the circulatory levels of Tregs and the pathological response of the breast cancers to NAC.


Subject(s)
Breast Neoplasms/drug therapy , CTLA-4 Antigen/metabolism , Chemotherapy, Adjuvant/methods , Forkhead Transcription Factors/metabolism , Neoadjuvant Therapy/methods , T-Lymphocytes, Regulatory/cytology , Breast Neoplasms/surgery , Combined Modality Therapy/methods , Female , Gene Expression Regulation, Neoplastic , Humans , Magnetic Resonance Imaging , Mammography , Phenotype , RNA, Messenger/metabolism , Th1 Cells/cytology , Th17 Cells/cytology , Th2 Cells/cytology
6.
Eur J Cancer ; 45(7): 1274-1281, 2009 May.
Article in English | MEDLINE | ID: mdl-19171478

ABSTRACT

Neoadjuvant chemotherapy is used in women who have large or locally advanced breast cancers. However, up to 70% of women who receive neoadjuvant chemotherapy fail to achieve a complete pathological response in their primary tumour (a surrogate marker of long-term survival). Five proteins, previously identified to be linked with chemoresistance in our in vitro experiments, were identified histochemically in pre-treatment core needle biopsies from 40 women with large or locally advanced breast cancers. Immunohistochemical staining with the five proteins showed no single protein to be a predictor of response to chemotherapy. However, pre-treatment breast cancer specimens that were annexin-A2 positive but annexin-A1 negative correlated with a poor pathological response (p=0.04, Fisher's exact test). The mechanisms by which annexins confer chemoresistance have not been identified, but may be due to inhibition of apoptosis. Annexin-A1 has been shown to enhance apoptosis, whilst annexin-A2, by contrast, inhibits apoptosis.


Subject(s)
Annexin A1/analysis , Annexin A2/analysis , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Carcinoma, Ductal, Breast/chemistry , Adult , Aged , Biopsy, Needle , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Chemotherapy, Adjuvant , Chi-Square Distribution , Female , Humans , Immunohistochemistry , Middle Aged , Neoadjuvant Therapy , Prognosis , Receptors, Estrogen/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...