Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38928129

ABSTRACT

Peripheral blood CD8+ T lymphocytes play a crucial role in cell-mediated immunity and tumor-related immune responses in breast cancer. In this study, label-free quantification analysis and gene set enrichment analysis (GSEA) of CD8+ T lymphocytes in the peripheral blood of benign patients and patients with different breast cancer (BC) subtypes, i.e., luminal A, luminal B, and triple-negative breast cancer (TNBC), were performed using nano-UHPLC and Orbitrap mass spectrometry. Differential protein expression in CD8+ T lymphocytes revealed significant downregulation (log2 FC ≥ 0.38 or ≤-0.38, adj. p < 0.05), particularly in proteins involved in cytotoxicity, cytolysis, and proteolysis, such as granzymes (GZMs) and perforin 1 (PRF1). This downregulation was observed in the benign group (GZMH, GZMM, and PRF1) and luminal B (GZMA, GZMH) subtypes, whereas granzyme K (GZMK) was upregulated in TNBC in comparison to healthy controls. The RNA degradation pathway was significantly downregulated (p < 0.05, normalized enrichment score (NES) from -1.47 to -1.80) across all BC subtypes, suggesting a potential mechanism for regulating gene expression during T cell activation. Also, the Sm-like proteins (LSM2, LSM3, and LSM5) were significantly downregulated in the RNA degradation pathway. Proteomic analysis of CD8+ T lymphocytes in peripheral blood across different breast cancer subtypes provides a comprehensive view of the molecular mechanisms of the systemic immune response that can significantly contribute to advancements in the diagnosis, treatment, and prognosis of this disease.


Subject(s)
Breast Neoplasms , CD8-Positive T-Lymphocytes , Granzymes , Humans , Female , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/blood , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Middle Aged , Granzymes/metabolism , Granzymes/genetics , Granzymes/blood , Adult , Perforin/metabolism , Perforin/genetics , Aged , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic
2.
Breast Cancer Res ; 26(1): 97, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858721

ABSTRACT

BACKGROUND: Tumor immune infiltration and peripheral blood immune signatures have prognostic and predictive value in breast cancer. Whether distinct peripheral blood immune phenotypes are associated with response to neoadjuvant chemotherapy (NAC) remains understudied. METHODS: Peripheral blood mononuclear cells from 126 breast cancer patients enrolled in a prospective clinical trial (NCT02022202) were analyzed using Cytometry by time-of-flight with a panel of 29 immune cell surface protein markers. Kruskal-Wallis tests or Wilcoxon rank-sum tests were used to evaluate differences in immune cell subpopulations according to breast cancer subtype and response to NAC. RESULTS: There were 122 evaluable samples: 47 (38.5%) from patients with hormone receptor-positive, 39 (32%) triple-negative (TNBC), and 36 (29.5%) HER2-positive breast cancer. The relative abundances of pre-treatment peripheral blood T, B, myeloid, NK, and unclassified cells did not differ according to breast cancer subtype. In TNBC, higher pre-treatment myeloid cells were associated with lower pathologic complete response (pCR) rates. In hormone receptor-positive breast cancer, lower pre-treatment CD8 + naïve and CD4 + effector memory cells re-expressing CD45RA (TEMRA) T cells were associated with more extensive residual disease after NAC. In HER2 + breast cancer, the peripheral blood immune phenotype did not differ according to NAC response. CONCLUSIONS: Pre-treatment peripheral blood immune cell populations (myeloid in TNBC; CD8 + naïve T cells and CD4 + TEMRA cells in luminal breast cancer) were associated with response to NAC in early-stage TNBC and hormone receptor-positive breast cancers, but not in HER2 + breast cancer. TRIAL REGISTRATION: NCT02022202 . Registered 20 December 2013.


Subject(s)
Breast Neoplasms , Immunophenotyping , Neoadjuvant Therapy , Humans , Female , Neoadjuvant Therapy/methods , Middle Aged , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/blood , Breast Neoplasms/pathology , Adult , Aged , Receptor, ErbB-2/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukocytes, Mononuclear/metabolism , Biomarkers, Tumor/blood , Prognosis , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/pathology , Prospective Studies , Treatment Outcome , Chemotherapy, Adjuvant/methods
3.
Comput Biol Med ; 176: 108537, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744008

ABSTRACT

BACKGROUND: Anti-PD-1/PD-L1 treatment has achieved durable responses in TNBC patients, whereas a fraction of them showed non-sensitivity to the treatment and the mechanism is still unclear. METHODS: Pre- and post-treatment plasma samples from triple negative breast cancer (TNBC) patients treated with immunotherapy were measured by tandem mass tag (TMT) mass spectrometry. Public proteome data of lung cancer and melanoma treated with immunotherapy were employed to validate the findings. Blood and tissue single-cell RNA sequencing (scRNA-seq) data of TNBC patients treated with or without immunotherapy were analyzed to identify the derivations of plasma proteins. RNA-seq data from IMvigor210 and other cancer types were used to validate plasma proteins in predicting response to immunotherapy. RESULTS: A random forest model constructed by FAP, LRG1, LBP and COMP could well predict the response to immunotherapy. The activation of complement cascade was observed in responders, whereas FAP and COMP showed a higher abundance in non-responders and negative correlated with the activation of complements. scRNA-seq and bulk RNA-seq analysis suggested that FAP, COMP and complements were derived from fibroblasts of tumor tissues. CONCLUSIONS: We constructe an effective plasma proteomic model in predicting response to immunotherapy, and find that FAP+ and COMP+ fibroblasts are potential targets for reversing immunotherapy resistance.


Subject(s)
Immunotherapy , Proteomics , Single-Cell Analysis , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Female , Immunotherapy/methods , Single-Cell Analysis/methods , Proteomics/methods , B7-H1 Antigen/blood , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Transcriptome , Immune Checkpoint Inhibitors/therapeutic use , Gene Expression Profiling , Proteome
4.
Clin Cancer Res ; 30(11): 2444-2451, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38470545

ABSTRACT

PURPOSE: We previously demonstrated the clinical significance of circulating tumor DNA (ctDNA) in patients with HER2-negative breast cancer receiving neoadjuvant chemotherapy (NAC). Here, we compared its predictive and prognostic value with cell-free DNA (cfDNA) concentration measured in the same samples from the same patients. EXPERIMENTAL DESIGN: 145 patients with hormone receptor (HR)-positive/HER2-negative and 138 triple-negative breast cancer (TNBC) with ctDNA data from a previous study were included in the analysis. Associations of serial cfDNA concentration with residual cancer burden (RCB) and distant recurrence-free survival (DRFS) were examined. RESULTS: In TNBC, we observed a modest negative correlation between cfDNA concentration 3 weeks after treatment initiation and RCB, but none of the other timepoints showed significant correlation. In contrast, ctDNA was significantly positively correlated with RCB at all timepoints (all R > 0.3 and P < 0.05). In the HR-positive/HER2-negative group, cfDNA concentration did not associate with response to NAC, but survival analysis showed that high cfDNA shedders at pretreatment had a significantly worse DRFS than low shedders (hazard ratio, 2.12; P = 0.037). In TNBC, the difference in survival between high versus low cfDNA shedders at all timepoints was not statistically significant. In contrast, as previously reported, ctDNA at all timepoints was significantly correlated with DRFS in both subtypes. CONCLUSIONS: In TNBC, cfDNA concentrations during therapy were not strongly correlated with response or prognosis. In the HR-positive/HER2-negative group, pretreatment cfDNA concentration was prognostic for DRFS. Overall, the predictive and prognostic value of cfDNA concentration was more limited than that of ctDNA.


Subject(s)
Biomarkers, Tumor , Cell-Free Nucleic Acids , Circulating Tumor DNA , Neoadjuvant Therapy , Neoplasm Recurrence, Local , Receptor, ErbB-2 , Triple Negative Breast Neoplasms , Humans , Female , Neoadjuvant Therapy/methods , Biomarkers, Tumor/blood , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Middle Aged , Prognosis , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/blood , Adult , Aged , Cell-Free Nucleic Acids/blood , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/blood , Treatment Outcome
5.
Breast Cancer Res Treat ; 205(3): 555-565, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38472594

ABSTRACT

PURPOSE: Recent evidence suggests that age-accumulated methylmalonic acid (MMA) promotes breast cancer progression in mice. This study aims to investigate the association between baseline serum MMA concentrations in patients with breast cancer and the development of subsequent distant metastases. METHODS: We included 32 patients with early Luminal B-like breast cancer (LumB, median age 62.4y) and 52 patients with early triple-negative breast cancer (TNBC, median age 50.5y) who developed distant metastases within 5 years. They were matched to an equal number of early breast cancer patients (median age 62.2y for LumB and 50.5y for TNBC) who did not develop distant metastases with at least 5 years of follow-up. RESULTS: Baseline serum MMA levels at breast cancer diagnosis showed a positive correlation with age (P < 0.001) and a negative correlation with renal function and vitamin B12 (all P < 0.02), but no statistical association was found with BMI or tumor stage (P > 0.6). Between matched pairs, no significant difference was observed in MMA levels, after adjusting for kidney function and age (P = 0.19). Additionally, in a mouse model, a significant decline in MMA levels was observed in the tumor-bearing group compared to the group without tumors before and after tumor establishment or at identical times for the control group (P = 0.03). CONCLUSION: Baseline serum MMA levels in patients with breast cancer are not correlated with secondary distant metastasis. Evidence in the mouse model suggests that the presence of a tumor perturbates MMA levels.


Subject(s)
Breast Neoplasms , Methylmalonic Acid , Neoplasm Metastasis , Humans , Female , Methylmalonic Acid/blood , Animals , Middle Aged , Mice , Breast Neoplasms/blood , Breast Neoplasms/pathology , Breast Neoplasms/diagnosis , Aged , Adult , Aging/blood , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/diagnosis , Neoplasm Staging , Age Factors
6.
Oncologist ; 29(5): e635-e642, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38431781

ABSTRACT

BACKGROUND: Our previous work indicated that the addition of lobaplatin to combined therapy with taxane and anthracycline can improve the pathological complete response rate of neoadjuvant therapy for triple-negative breast cancer (TNBC) and lengthen long-term survival significantly, but the therapeutic markers of this regimen are unclear. METHODS: Eighty-three patients who met the inclusion criteria were included in this post hoc analysis. We analyzed the association between platelet-to-lymphocyte ratio (PLR) and neutrophil-to-lymphocyte ratio (NLR) before neoadjuvant chemotherapy with the efficacy and prognosis after treatment with docetaxel, epirubicin, and lobaplatin neoadjuvant chemotherapy regimen. χ2 test and Cox regression were used to analyze the association between PLR and NLR with total pathologic complete response (tpCR), as well as the association between PLR and NLR with event-free survival (EFS) and overall survival (OS), respectively. RESULTS: The tpCR rate in the PLR- group was 49.0% (25/51), which was significantly higher than that in the PLR+ group (25.0% [8/32], P = .032). The tpCR rate in the NLR- group was 49.1% (26/53), which was significantly higher than that in the NLR+ group (23.3% [7/30], P = .024). The tpCR rate of the PLR-NLR- (PLR- and NLR-) group was 53.7% (22/41), which was significantly higher than that of the PLR+/NLR+ (PLR+ or/and NLR+) group (26.1% [11/42]; P = .012). EFS and OS in the NLR+ group were significantly shorter than those in the NLR- group (P = .028 for EFS; P = .047 for OS). Patients in the PLR-NLR- group had a longer EFS than those in the PLR+/NLR+ group (P = .002). CONCLUSION: PLR and NLR could be used to predict the efficacy of neoadjuvant therapy with the taxane, anthracycline, and lobaplatin regimen for patients with TNBC, as patients who had lower PLR and NLR values had a higher tpCR rate and a better long-term prognosis.


Subject(s)
Cyclobutanes , Neoadjuvant Therapy , Organoplatinum Compounds , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/mortality , Female , Neoadjuvant Therapy/methods , Prognosis , Middle Aged , Cyclobutanes/pharmacology , Cyclobutanes/therapeutic use , Cyclobutanes/administration & dosage , Organoplatinum Compounds/therapeutic use , Organoplatinum Compounds/pharmacology , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aged , Neutrophils/metabolism , Biomarkers, Tumor/blood , Lymphocytes/metabolism , Blood Platelets/pathology , Retrospective Studies
7.
Ann Oncol ; 34(2): 200-211, 2023 02.
Article in English | MEDLINE | ID: mdl-36423745

ABSTRACT

BACKGROUND: Post-treatment detection of circulating tumour DNA (ctDNA) in early-stage triple-negative breast cancer (TNBC) patients predicts high risk of relapse. c-TRAK TN assessed the utility of prospective ctDNA surveillance in TNBC and the activity of pembrolizumab in patients with ctDNA detected [ctDNA positive (ctDNA+)]. PATIENTS AND METHODS: c-TRAK TN, a multicentre phase II trial, with integrated prospective ctDNA surveillance by digital PCR, enrolled patients with early-stage TNBC and residual disease following neoadjuvant chemotherapy, or stage II/III with adjuvant chemotherapy. ctDNA surveillance comprised three-monthly blood sampling to 12 months (18 months if samples were missed due to coronavirus disease), and ctDNA+ patients were randomised 2 : 1 to intervention : observation. ctDNA results were blinded unless patients were allocated to intervention, when staging scans were done and those free of recurrence were offered pembrolizumab. A protocol amendment (16 September 2020) closed the observation group; all subsequent ctDNA+ patients were allocated to intervention. Co-primary endpoints were (i) ctDNA detection rate and (ii) sustained ctDNA clearance rate on pembrolizumab (NCT03145961). RESULTS: Two hundred and eight patients registered between 30 January 2018 and 06 December 2019, 185 had tumour sequenced, 171 (92.4%) had trackable mutations, and 161 entered ctDNA surveillance. Rate of ctDNA detection by 12 months was 27.3% (44/161, 95% confidence interval 20.6% to 34.9%). Seven patients relapsed without prior ctDNA detection. Forty-five patients entered the therapeutic component (intervention n = 31; observation n = 14; one observation patient was re-allocated to intervention following protocol amendment). Of patients allocated to intervention, 72% (23/32) had metastases on staging at the time of ctDNA+, and 4 patients declined pembrolizumab. Of the five patients who commenced pembrolizumab, none achieved sustained ctDNA clearance. CONCLUSIONS: c-TRAK TN is the first prospective study to assess whether ctDNA assays have clinical utility in guiding therapy in TNBC. Patients had a high rate of metastatic disease on ctDNA detection. Findings have implications for future trial design, emphasising the importance of commencing ctDNA testing early, with more sensitive and/or frequent ctDNA testing regimes.


Subject(s)
Antineoplastic Agents, Immunological , Circulating Tumor DNA , Neoplasm, Residual , Triple Negative Breast Neoplasms , Humans , Biomarkers, Tumor/blood , Mutation , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Prospective Studies , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Neoplasm, Residual/blood , Neoplasm, Residual/diagnosis , Neoplasm, Residual/drug therapy , Neoplasm, Residual/genetics , Antineoplastic Agents, Immunological/therapeutic use , Circulating Tumor DNA/blood
8.
Pathol Res Pract ; 231: 153810, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35182891

ABSTRACT

PURPOSE: Triple Negative Breast Cancer (TNBC) is the malignant tumor with the fastest progression rate in breast cancer. LncRNAs are widely involved in various biological characteristics of tumor. The purpose of this study was to mine LncRNAs that can be used to diagnose and evaluate the prognosis of TNBC. METHODS: Base on TCGA dataset, we used three R language packages to analyze the differentially expressed (DE) lncRNAs in TNBC. Survival analysis and ROC curve analysis were conducted to estimate the potential diagnostic and prognostic value of LncRNAs for TNBC. Furthermore, CCK-8 and Transwell assays were used to assess the effects of LncRNA on MDA-MB-231 cells proliferation and migration. Additionally, targets mRNAs of candidate LncRNA were predicted by co-expression analysis and multiple target gene prediction databases, then KEGG pathway and GO analysis were conducted using DAVID online tool. RESULTS: 6165 DERNAs and 1258 DElncRNAs were obtained. 40 LncRNAs were significantly correlated with the survival time of TNBC patients. Among them, HAGLROS has the highest HR value. ROC curve analysis also showed that HAGLROS had high sensitivity and specificity. Further in vitro experiments showed that downregulation of HAGLROS inhibited the proliferation and migration of MDA-MB-231 cells. Moreover, by conducting bioinformatics analysis, we found that these target genes of HAGLROS were involved in regulating five signaling pathways. Mechanistic investigations demonstrated that HAGLROS might regulate the expression of PAX5 through miR-330-5p, the effects of miR-330-5p in MDA-MB-231 cells were also analyzed. CONCLUSION: Our results showed that HAGLROS was significantly overexpressed in TNBC, and high HAGLROS expression predicted poor overall survival. Downregulation of HAGLROS could inhibite the proliferation and migration of MDA-MB-231 cell by regulating PAX5 expression through miR-330-5p.


Subject(s)
RNA, Long Noncoding/analysis , Triple Negative Breast Neoplasms/genetics , Cell Proliferation/genetics , Humans , Prognosis , RNA, Long Noncoding/blood , Triple Negative Breast Neoplasms/blood
9.
Cells ; 11(2)2022 01 13.
Article in English | MEDLINE | ID: mdl-35053375

ABSTRACT

Although some therapies are available for regular breast cancers, there are very few options for triple-negative breast cancer (TNBC). Here, we demonstrated that serum level of IL-12p40 monomer (p40) was much higher in breast cancer patients than healthy controls. On the other hand, levels of IL-12, IL-23 and p40 homodimer (p402) were lower in serum of breast cancer patients as compared to healthy controls. Similarly, human TNBC cells produced greater level of p40 than p402. The level of p40 was also larger than p402 in serum of a patient-derived xenograft (PDX) mouse model. Accordingly, neutralization of p40 by p40 mAb induced death of human TNBC cells and tumor shrinkage in PDX mice. While investigating the mechanism, we found that neutralization of p40 led to upregulation of human CD4+IFNγ+ and CD8+IFNγ+ T cell populations, thereby increasing the level of human IFNγ and decreasing the level of human IL-10 in PDX mice. Finally, we demonstrated the infiltration of human cytotoxic T cells, switching of tumor-associated macrophage M2 (TAM2) to TAM1 and suppression of transforming growth factor ß (TGFß) in tumor tissues of p40 mAb-treated PDX mice. Our studies identify a possible new immunotherapy for TNBC in which p40 mAb inhibits tumor growth in PDX mice.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Interleukin-12 Subunit p40/immunology , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Adaptive Immunity/drug effects , Animals , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Immunotherapy , Interferon-gamma/metabolism , Interleukin-12/blood , Interleukin-12/metabolism , Interleukin-12 Subunit p40/blood , Interleukin-23/blood , Interleukin-23/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred NOD , Mice, SCID , Neutralization Tests , Spleen/metabolism , Triple Negative Breast Neoplasms/blood , Up-Regulation
11.
Immunotherapy ; 14(4): 189-199, 2022 03.
Article in English | MEDLINE | ID: mdl-34984928

ABSTRACT

Introduction: There is evidence for an association between peripheral blood eosinophil count (PBEC) and response to cancer immunotherapy; however, such data is limited in metastatic triple-negative breast cancer (mTNBC). Patients & methods: This report presents patients (n = 14) who received a combination of durvalumab and paclitaxel for mTNBC (NCT02628132). Results: There was a statistically significant correlation (p = 0.028) between an increase in PBEC (>300/mm3) during treatment and response to the combination therapy. Survival analysis showed a statistically significant association between progression-free survival and increased PBEC, after therapy (p = 0.005). A similar trend existed for overall survival, although it did not reach statistical significance (p = 0.167). Conclusion: This is the first study to report on eosinophilia in mTNBC treated with chemoimmunotherapy and supports a role for eosinophils in immunotherapy for mTNBC.


Plain language summary Previous reports have shown that an increase in peripheral blood eosinophil count is associated with a good response to cancer immunotherapy; however, data on this association is limited in a subtype of breast cancer called metastatic triple-negative breast cancer (mTNBC). The eosinophil count in patients who received the combination of an immunotherapeutic agent, durvalumab and a chemotherapeutic agent, paclitaxel, in mTNBC was assessed. There was a statistically significant association between an increase in eosinophils during treatment and the response to therapy. The progression of the disease was slower or less likely to occur in patients with increased eosinophils. This study supports the role of eosinophils in immunotherapy for mTNBC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/blood , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Eosinophils/drug effects , Immunotherapy/methods , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/drug therapy , Adult , Eosinophils/metabolism , Female , Humans , Leukocyte Count , Middle Aged , Progression-Free Survival , Treatment Outcome
12.
Front Immunol ; 12: 753477, 2021.
Article in English | MEDLINE | ID: mdl-34777365

ABSTRACT

Slit2 exerts antitumor effects in various cancers; however, the underlying mechanism, especially its role in regulating the immune, especially in the bone marrow niche, system is still unknown. Elucidating the behavior of macrophages in tumor progression can potentially improve immunotherapy. Using a spontaneous mammary tumor virus promoter-polyoma middle T antigen (PyMT) breast cancer mouse model, we observed that Slit2 increased the abundance of antitumor M1 macrophage in the bone marrow upon differentiation in vitro. Moreover, myeloablated PyMT mice injected with Slit2-treated bone marrow allografts showed a marked reduction in tumor growth, with enhanced recruitment of M1 macrophage in their tumor stroma. Mechanistic studies revealed that Slit2 significantly enhanced glycolysis and reduced fatty acid oxidation in bone marrow-derived macrophages (BMDMs). Slit2 treatment also altered mitochondrial respiration metabolites in macrophages isolated from healthy human blood that were treated with plasma from breast cancer patients. Overall, this study, for the first time, shows that Slit2 increases BMDM polarization toward antitumor phenotype by modulating immune-metabolism. Furthermore, this study provides evidence that soluble Slit2 could be developed as novel therapeutic strategy to enhance antitumor immune response.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Macrophage Activation/drug effects , Macrophages/drug effects , Mammary Neoplasms, Experimental/therapy , Metabolome/drug effects , Nerve Tissue Proteins/physiology , Adult , Aged , Animals , Antigens, Polyomavirus Transforming/genetics , Culture Media, Conditioned , Female , Glycolysis/drug effects , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Lipopolysaccharide Receptors/analysis , Macrophages/immunology , Macrophages/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Transgenic , Middle Aged , Monocytes/drug effects , Monocytes/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Radiation Chimera , TOR Serine-Threonine Kinases/physiology , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/chemistry , Tumor Burden
13.
Clin Epigenetics ; 13(1): 207, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34789319

ABSTRACT

BACKGROUND: A shift in the proportions of blood immune cells is a hallmark of cancer development. Here, we investigated whether methylation-derived immune cell type ratios and methylation-derived neutrophil-to-lymphocyte ratios (mdNLRs) are associated with triple-negative breast cancer (TNBC). METHODS: Leukocyte subtype-specific unmethylated/methylated CpG sites were selected, and methylation levels at these sites were used as proxies for immune cell type proportions and mdNLR estimation in 231 TNBC cases and 231 age-matched controls. Data were validated using the Houseman deconvolution method. Additionally, the natural killer (NK) cell ratio was measured in a prospective sample set of 146 TNBC cases and 146 age-matched controls. RESULTS: The mdNLRs were higher in TNBC cases compared with controls and associated with TNBC (odds ratio (OR) range (2.66-4.29), all Padj. < 1e-04). A higher neutrophil ratio and lower ratios of NK cells, CD4 + T cells, CD8 + T cells, monocytes, and B cells were associated with TNBC. The strongest association was observed with decreased NK cell ratio (OR range (1.28-1.42), all Padj. < 1e-04). The NK cell ratio was also significantly lower in pre-diagnostic samples of TNBC cases compared with controls (P = 0.019). CONCLUSION: This immunomethylomic study shows that a shift in the ratios/proportions of leukocyte subtypes is associated with TNBC, with decreased NK cell showing the strongest association. These findings improve our knowledge of the role of the immune system in TNBC and point to the possibility of using NK cell level as a non-invasive molecular marker for TNBC risk assessment, early detection, and prevention.


Subject(s)
Leukocyte Count/statistics & numerical data , Triple Negative Breast Neoplasms/genetics , Adult , Case-Control Studies , DNA Methylation/genetics , DNA Methylation/immunology , Epigenomics/methods , Epigenomics/statistics & numerical data , Female , Humans , Leukocyte Count/classification , Leukocyte Count/methods , Logistic Models , Middle Aged , Odds Ratio , Proportional Hazards Models , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/immunology
14.
Mol Cell Proteomics ; 20: 100121, 2021.
Article in English | MEDLINE | ID: mdl-34265469

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer. Patients with TNBC have poor overall survival because of limited molecular therapeutic targets. Recently, exosomes have been recognized as key mediators in cancer progression, but the molecular components and function of TNBC-derived exosomes remain unknown. The main goal of this study was to reveal the proteomic landscape of serum exosomes derived from ten patients with TNBC and 17 healthy donors to identify potential therapeutic targets. Using a tandem mass tag-based quantitative proteomics approach, we characterized the proteomes of individual patient-derived serum exosomes, identified exosomal protein signatures specific to patients with TNBC, and filtered out differentially expressed proteins. Most importantly, we found that the tetraspanin CD151 expression levels in TNBC-derived serum exosomes were significantly higher than those exosomes from healthy subjects, and we validated our findings with samples from 16 additional donors. Furthermore, utilizing quantitative proteomics approach to reveal the proteomes of CD151-deleted exosomes and cells, we found that exosomal CD151 facilitated secretion of ribosomal proteins via exosomes while inhibiting exosome secretion of complement proteins. Moreover, we proved that CD151-deleted exosomes significantly decreased the migration and invasion of TNBC cells. This is the first comparative study of the proteomes of TNBC patient-derived and CD151-deleted exosomes. Our findings indicate that profiling of TNBC-derived exosomal proteins is a useful tool to extend our understanding of TNBC, and exosomal CD151 may be a potential therapeutic target for TNBC.


Subject(s)
Exosomes/metabolism , Proteome/metabolism , Tetraspanin 24/metabolism , Triple Negative Breast Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Middle Aged , Protein Interaction Maps , Tetraspanin 24/genetics , Triple Negative Breast Neoplasms/blood
15.
Breast Dis ; 40(4): 241-250, 2021.
Article in English | MEDLINE | ID: mdl-34092580

ABSTRACT

OBJECTIVE: Molecular markers for the detection of breast cancer and its different types, grades, and stages lack enough sensitivity and specificity. This study evaluates the expression of miRNAs 9 and 342 in sera of different types, grades, and stages of BC. Moreover, the assessment of their sensitivity, specificity, diagnostic, and prognostic role in detecting different types of BC. METHODS: Blood was collected from 200 females outpatients, divided into five groups each 40 subjects: control, benign breast tumor, estrogen receptor (ER+)/progesterone receptor (PR+) BC, human epidermal growth factor receptor (HER+) BC, and triple-negative BC. BC subjects were further subdivided according to grade and stage. Expressions of miRNAs 9 and 342 were measured for all subjects by real-time polymerase chain reaction (RT-PCR). RESULTS: Results showed that serum expression of both miRNAs 9 and 342 can be used for the diagnosis of different types of BC. Their expression can be used to significantly differentiate between different grades and stages of BC. MiRNAs 9 and 342 showed high sensitivity of 92.5% and specificity of (81.2 and 88.7%), respectively, for triple-negative BC. CONCLUSION: The expressions of miRNAs 9 and 342 provide potential roles as serological biomarkers for the diagnosis and prognosis of different types, grades, and stages of BC.


Subject(s)
Breast Neoplasms/blood , MicroRNAs/blood , Adult , Biomarkers, Tumor , Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Female , Humans , Middle Aged , Prognosis , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Sensitivity and Specificity , Triple Negative Breast Neoplasms/blood
16.
J Cell Mol Med ; 25(16): 7602-7607, 2021 08.
Article in English | MEDLINE | ID: mdl-33949761

ABSTRACT

Exosomal lncRNAs secreted by cancer cells can serve as potential biomarkers in the diagnosis and prognosis of various tumours. Here, we are committed to explore the diagnostic and prognostic value of serum exosomal XIST secreted by tumour cells to predict recurrence in patients with triple-negative breast cancer (TNBC). Significant increments in XIST and exo-XIST from tumour tissues and blood serum were found in reoccurring TNBC patients by comparison with non-recurrences. Levels of serum exo-XIST were only significantly increased in TNBC recurrence and no association with other clinicopathological parameters. Additionally, serum exo-XIST levels could be served as an assessment of change in the load of triple-negative breast cancer. Expressions of exo-XIST were markedly decreased after resection of the primary breast tumours and obviously elevated at the time of recurrence. Finally, an obvious association was identified between serum exo-XIST levels and a poorer overall survival (OS) in TNBC patients. Levels of serum exo-XIST may serve as a diagnostic and prognostic biomarker to predict the recurrent TNBC-loading status.


Subject(s)
Biomarkers, Tumor/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Neoplasm Recurrence, Local/pathology , RNA, Long Noncoding/genetics , Triple Negative Breast Neoplasms/pathology , Biomarkers, Tumor/blood , Case-Control Studies , Exosome Multienzyme Ribonuclease Complex/blood , Humans , Middle Aged , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/genetics , Prognosis , RNA, Long Noncoding/blood , ROC Curve , Survival Rate , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/genetics
17.
Theranostics ; 11(13): 6445-6460, 2021.
Article in English | MEDLINE | ID: mdl-33995667

ABSTRACT

Background: Neoadjuvant chemotherapy is relevant to the formation of thromboembolism and secondary neoplasms in triple-negative breast cancer (TNBC). Chemotherapy-induced breast cancer cell-derived microparticles (BCMPs) may have important thrombogenic and pro-metastatic effects on platelets and endothelium, which may be related to the expression and distribution of phosphatidylserine (PS). However, investigating these interactions is challenging due to technical limitations. Methods: A study was conducted in 20 healthy individuals and 18 patients who had been recently diagnosed with TNBC and were undergoing neoadjuvant chemotherapy with doxorubicin and cyclophosphamide. BCMPs were isolated from patient blood samples and doxorubicin-treated breast cancer cell lines. Their structure and morphology were studied by electron microscopy and antigen levels were measured by fluorescence-activated cell sorting. In an inhibition assay, isolated BCMPs were pretreated with lactadherin or tissue factor antibodies. Platelets isolated from healthy subjects were treated with BCMPs and coagulation time, fibrin formation, and expression of intrinsic/extrinsic factor Xase (FXa) and thrombin were evaluated. The effects of BCMPs on endothelial thrombogenicity and integrity were assessed by confocal microscopy, electron microscopy, measurement of intrinsic/extrinsic FXa, prothrombinase assay, and transwell permeability assay. Results: Neoadjuvant chemotherapy significantly increased the expression of PS+ BCMPs in patient plasma. Its expression was associated with a rapid increase in procoagulant activity. Treatment with lactadherin, a PS-binding scavenging molecule, markedly reduced the adhesion of BCMPs and abolished their procoagulant activity, but this was not observed with tissue factor antibody treatment. Intravenous injection of BCMPs in mice induced a significant hypercoagulable state, reducing the extent of plasma fibrinogen and promoting the appearance of new thrombus. Cancer cells incubated with doxorubicin released large numbers of PS+ BCMPs, which stimulated and transformed endothelial cells into a procoagulant phenotype and increased the aggregation and activation of platelets. Moreover, cancer cells exploited this BCMP-induced endothelial leakiness and showed promoted metastasis. Pretreatment with lactadherin increased uptake of both PS+ BCMPs and cancer cells by endothelial cells and limited the transendothelial migration of cancer cells. Conclusion: Lactadherin, a biosensor that we developed, was used to study the extracellular vesicle distribution of PS, which revealed a novel PS+ BCMPs administrative axis that initiated a local coagulation cascade and facilitated metastatic colonization of circulating cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cell-Derived Microparticles/physiology , Membrane Lipids/analysis , Neoadjuvant Therapy/adverse effects , Phosphatidylserines/analysis , Thrombophilia/etiology , Transendothelial and Transepithelial Migration , Triple Negative Breast Neoplasms/pathology , Aged , Animals , Antibodies/immunology , Antigens, Surface/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Coagulation Factors/analysis , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Endothelium, Vascular/pathology , Female , Fibrinolysis , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Middle Aged , Milk Proteins/pharmacology , Thromboplastin/immunology , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/drug therapy
18.
J Immunother ; 44(7): 264-275, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33928928

ABSTRACT

LY3381916 is an orally available, highly selective, potent inhibitor of indoleamine 2,3-dioxygenase 1. This study explored the safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity of LY3381916 monotherapy and in combination with a programmed death-ligand 1 (PD-L1) inhibitor (LY3300054) in patients with advanced solid tumors. During dose escalation, patients received escalating doses of LY3381916 at 60-600 mg once daily (qd) and 240 mg twice daily in monotherapy (n=21) and in combination with PD-L1 inhibitor at 700 mg every 2 weeks (n=21). A modified toxicity probability interval method was used to guide dose escalation. Dose-limiting toxicities occurred in 3 patients; 1 at LY3381916 240 mg twice daily (alanine aminotransferase/aspartate aminotransferase increase and systemic inflammatory response syndrome) and 2 at LY3381916 240 mg qd in combination with PD-L1 inhibitor (fatigue and immune-related hepatitis). LY3381916, at the recommended phase II dose, 240 mg qd, in combination with PD-L1 inhibitor, produced maximal inhibition of indoleamine 2,3-dioxygenase 1 activity in plasma and tumor tissue, and led to an increase of CD8 T cells in tumor tissue. In the combination dose expansion cohorts, 14 triple-negative breast cancer and 4 non-small cell lung cancer patients were enrolled. Treatment-related liver toxicity (grade ≥2 alanine aminotransferase/aspartate aminotransferase increase or immune-related hepatitis) was the most prominent adverse event in triple-negative breast cancer patients (n=5, 35.7%). Best response was stable disease. These preliminary data suggest an alternative dose level of LY3381916 is needed for the combination with PD-L1 inhibitor. The combination clinical activity was limited in this study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung , Indoleamine-Pyrrole 2,3,-Dioxygenase , Lung Neoplasms , Triple Negative Breast Neoplasms , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/blood , Kynurenine/blood , Lung Neoplasms/blood , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism
19.
Genes (Basel) ; 12(4)2021 04 09.
Article in English | MEDLINE | ID: mdl-33918859

ABSTRACT

Resistance to therapy is a persistent problem that leads to mortality in breast cancer, particularly triple-negative breast cancer (TNBC). MiRNAs have become a focus of investigation as tissue-specific regulators of gene networks related to drug resistance. Circulating miRNAs are readily accessible non-invasive potential biomarkers for TNBC diagnosis, prognosis, and drug-response. Our aim was to use systems biology, meta-analysis, and network approaches to delineate the drug resistance pathways and clinical outcomes associated with circulating miRNAs in TNBC patients. MiRNA expression analysis was used to investigate differentially regulated circulating miRNAs in TNBC patients, and integrated pathway regulation, gene ontology, and pharmacogenomic network analyses were used to identify target genes, miRNAs, and drug interaction networks. Herein, we identified significant differentially expressed circulating miRNAs in TNBC patients (miR-19a/b-3p, miR-25-3p, miR-22-3p, miR-210-3p, miR-93-5p, and miR-199a-3p) that regulate several molecular pathways (PAM (PI3K/Akt/mTOR), HIF-1, TNF, FoxO, Wnt, and JAK/STAT, PD-1/PD-L1 pathways and EGFR tyrosine kinase inhibitor resistance (TKIs)) involved in drug resistance. Through meta-analysis, we demonstrated an association of upregulated miR-93, miR-210, miR-19a, and miR-19b with poor overall survival outcomes in TNBC patients. These results identify miRNA-regulated mechanisms of drug resistance and potential targets for combination with chemotherapy to overcome drug resistance in TNBC. We demonstrate that integrated analysis of multi-dimensional data can unravel mechanisms of drug-resistance related to circulating miRNAs, particularly in TNBC. These circulating miRNAs may be useful as markers of drug response and resistance in the guidance of personalized medicine for TNBC.


Subject(s)
Biomarkers, Tumor/genetics , Circulating MicroRNA/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Pharmaceutical Preparations/administration & dosage , Triple Negative Breast Neoplasms/pathology , Adult , Biomarkers, Tumor/blood , Case-Control Studies , Female , Gene Expression Profiling , Humans , Network Meta-Analysis , Prognosis , Survival Rate , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
20.
Int J Mol Sci ; 22(7)2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33916440

ABSTRACT

Several lines of evidence suggest that altered adenosine deaminase (ADA) activity, especially its ADA2 iso-enzyme, is associated with malignant breast cancer (BC) development. Triple-negative breast cancer (TNBC) is currently the most challenging BC subtype due to its metastatic potential and recurrence. Herein, we analyzed the sources of ADA iso-enzymes in TNBC by investigating the effects of cell-to-cell interactions between TNBC cells, macrophages, lymphocytes, and endothelial cells. We also examined the potential relationship between ADA activity and cancer progression in TNBC patients. In vitro analyses demonstrated that the interactions of immune and endothelial cells with MDA-MB-231 triple negative BC cells modulated their extracellular adenosine metabolism pattern. However, they caused an increase in the ADA1 activity, and did not alter ADA2 activity in cancer cells. In turn, the co-culture of MDA-MB-231 cells with THP-1 monocyte/macrophages, Jurkat cells, and human lung microvascular endothelial cells (HULEC) caused the increase in ADA2 activity on THP-1 cells and ADA1 activity on Jurkat cells and HULEC. Clinical sample analysis revealed that TNBC patients had higher plasma ADA2 activities and lower ADA1/ADA2 ratio at advanced stages of cancer development than in the initial stages, while patients with hormone receptor positive, HER2 negative (HR+HER2-), and triple positive (HR+HER2+) breast cancers at the same stages showed opposite trends. TNBC patients also demonstrated positive associations between plasma ADA2 activity and pro-tumor M2 macrophage markers, as well as between ADA1 activity and endothelial dysfunction or inflammatory parameters. The analysis of TNBC patients, at 6 and 12 months following cancer treatment, did not showed significant changes in plasma ADA activities and macrophage polarization markers, which may be the cause of their therapeutic failure. We conclude that alterations in both ADA iso-enzymes can play a role in breast cancer development and progression by the modulation of extracellular adenosine-dependent pathways. Additionally, the changes in ADA2 activity that may contribute to the differentiation of macrophages into unfavorable pro-tumor M2 phenotype deserve special attention in TNBC.


Subject(s)
Adenosine Deaminase/blood , Biomarkers, Tumor/blood , Intercellular Signaling Peptides and Proteins/blood , Macrophages/enzymology , Triple Negative Breast Neoplasms/blood , Adult , Female , Humans , Jurkat Cells , Macrophages/pathology , Middle Aged , THP-1 Cells , Triple Negative Breast Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...