Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Clin Med ; 12(12)2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37373727

ABSTRACT

BACKGROUND: Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) is a well-established diagnostic procedure for evaluating hilar and mediastinal lymphadenopathies and is the gold standard for lung cancer diagnosis and staging. Recent studies assessed the effectiveness of the 19-G flex needle in obtaining larger EBUS-TBNA samples, and prospective small series gave similar results in terms of diagnostic yield when testing different gauge needles. The lack of homogeneity between series and the small sample size of some prospective cohorts poses a limit to the validity of those results. This prospective controlled study compared the 19-G flex and 22-G needles in terms of diagnostic yield. An objective laboratory method was used to count cells and compare the two needles' cytologic yields. MATERIAL: A prospective controlled study was conducted on 90 patients undergoing EBUS-TBNA for the diagnosis of hilar and mediastinal lymphadenopathies. The institutional ethic committee (IEO573) approved the study, and informed consent was obtained from all patients. RESULTS: A total of 90 patients were enrolled in this study, 84.4% of whom were diagnosed with malignancy and 15.6% with non-neoplastic disease. Sensitivity for malignancy was 93.4% (CI: 87.4-97.1%) for the 19-G needle and 92.6% (CI: 86.3-96.5%) for the 22-G needle (p = 0.80). The percentage of malignant cells in the cell block was 63.9% and 61.5% for the 22-G and 19-G needles, respectively. The cell count assessed by flow cytometry was 2071 cells/µL (IQR: 600,2265) with the 22-G needle and 2761 cells/µL (IQR: 505,3250) with the 19-G needle (p = 0.79). The malignant cell count was 0.05 × 103 cells/µL with the 22-G and 0.08 × 103 cells/µL with the 19-G needle (p = 0.70). There was no difference in the presence of tissue cores in the samples, and rapid on-site evaluation (ROSE) cellularity was comparable between the two needles. CONCLUSIONS: The 19-G flex EBUS-TBNA needle is comparable to the 22-G needle in terms of diagnostic yield for cyto-histological evaluation of hilar and mediastinal lymphadenopathies. There is no difference between the 19-G and 22-G needle cell counts evaluated by flow cytometry.

2.
J Clin Med ; 12(7)2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37048617

ABSTRACT

We have previously shown in triple-negative breast cancer (TNBC) models that a triple therapy (TT) including intermittent cyclophosphamide (C), vinorelbine (V), and anti-PD-1 activates antigen-presenting cells (APC) and generates stem like-T cells able to control local and metastatic tumor progression. In the present manuscript, we report the generation of a highly aggressive, anti-PD-1 resistant model of a high-grade, Myc-driven B-cell non-Hodgkin's lymphoma (NHL) that can be controlled in vivo by TT but not by other chemotherapeutic agents, including cytarabine (AraC), platinum (P), and doxorubicin (D). The immunological memory elicited in tumor-bearing mice by TT (but not by other treatments) can effectively control NHL re-challenge even at very high inoculum doses. TT re-shaped the landscape of circulating innate NK cells and adaptive immune cells, including B and T cells, and significantly reduced exhausted CD4+ and CD8+ TIM3+PD-1+ T cells in the spleens of treated mice.

3.
Cell Immunol ; 382: 104615, 2022 12.
Article in English | MEDLINE | ID: mdl-36228388

ABSTRACT

The role and regulation of innate immune cells is poorly understood in B-cell non-Hodgkin lymphoma (NHL). As natural killer (NK) cells, helper innate lymphoid cells (ILCs) are lymphocytes endowed with either anti- or pro-tumour activity and involved in inflammatory processes. In our ex vivo analysis of NK cells and ILCs from NHL patients, we observed that, in comparison to healthy donors (HD), the frequency of the cytotoxic subset of NK cells, the CD16+ NK, decreased in patients' peripheral blood. In general, circulating NK cells showed a pro-tumorigenic phenotype, while ILCs displayed a more activated/cytotoxic phenotype. Conversely, at the tumour site, in patients' lymph nodes, ILCs showed a low expression of granzyme.In vitromixed lymphocyte-tumour cell cultures with HD PBMCs and NHL cell lines demonstrated that ILC cytotoxic potential was lowered by the presence of tumour cells but, in the absence of T regulatory cells (Tregs), their cytolytic potential was recovered. Our data shed novel light on dysfunctional innate immunity in NHL. We suggest a new mechanism of tumour immuno-escape based on the reduction of cell cytotoxicity involving ILCs and likely controlled by Tregs.


Subject(s)
Antineoplastic Agents , Lymphoma, Non-Hodgkin , Neoplasms , Humans , Tumor Escape , Immunity, Innate , Lymphocytes , Killer Cells, Natural , Neoplasms/pathology , Lymphoma, Non-Hodgkin/pathology
4.
Cancers (Basel) ; 14(18)2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36139632

ABSTRACT

BACKGROUND: The success of targeted therapies in the treatment of pancreatic neuroendocrine tumors has emphasized the strategy of targeting angiogenesis and the PI3K/AKT/mTOR pathway. However, the major challenge in the targeted era remains the early identification of resistant tumors especially when the efficacy is rarely associated to a clear tumor shrinkage at by imaging assessment. METHODS: In this prospective study (NCT02305810) we investigated the predictive and prognostic role of soluble biomarkers of angiogenesis turnover (VEGF, bFGF, VEGFR2, TSP-1) circulating endothelial cells and progenitors, in 43 patients with metastatic panNET receiving everolimus. RESULTS: Among all tested biomarkers, we found a specific subpopulation of circulating cells, CD31+CD140b-, with a significantly increased tumor progression hazard for values less or equal to the first quartile. CONCLUSION: Our study suggested the evidence that circulating cells might be surrogate biomarkers of angiogenesis activity in patients treated with everolimus and their baseline levels can be correlated with survival. However, further studies are now needed to validate the role of these cells as surrogate markers for the selection of patients to be candidates for antiangiogenic treatments.

5.
Cell Death Discov ; 8(1): 106, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35260564

ABSTRACT

Breast cancer (BC) constitutes a major health problem worldwide, making it the most common malignancy in women. Current treatment options for BC depend primarily on histological type, molecular markers, clinical aggressiveness and stage of disease. Immunotherapy, such as αPD-1, have shown combinatorial clinical activity with chemotherapy in triple negative breast cancer (TNBC) delineating some therapeutic combinations as more effective than others. However, a clear overview of the main immune cell populations involved in these treatments has never been provided.Here, an assessment of the immune landscape in the tumor microenvironment (TME) of two TNBC mouse models has been performed using single-cell RNA sequencing technology. Specifically, immune cells were evaluated in untreated conditions and after treatments with chemotherapy or immunotherapy used as single agents or in combination. A decrease of Treg was found in treatments with in vivo efficacy as well as γδ T cells, which have a pro-tumoral activity in mice. Focusing on Cd8 T cells, across all the conditions, a general increase of exhausted-like Cd8 T cells was confirmed in pre-clinical treatments with low efficacy and an opposite trend was found for the proliferative Cd8 T cells. Regarding macrophages, M2-like cells were enriched in treatments with low efficacy while M1-like macrophages followed an opposite trend. For both models, similar proportions of B cells were detected with an increase of proliferative B cells in treatments involving cisplatin in combination with αPD-1. The fine-scale characterization of the immune TME in this work can lead to new insights on the diagnosis and treatment of TNBC.

6.
Cancer Res ; 81(3): 685-697, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33268528

ABSTRACT

Checkpoint inhibitors (CI) instigate anticancer immunity in many neoplastic diseases, albeit only in a fraction of patients. The clinical success of cyclophosphamide (C)-based haploidentical stem-cell transplants indicates that this drug may re-orchestrate the immune system. Using models of triple-negative breast cancer (TNBC) with different intratumoral immune contexture, we demonstrate that a combinatorial therapy of intermittent C, CI, and vinorelbine activates antigen-presenting cells (APC), and abrogates local and metastatic tumor growth by a T-cell-related effect. Single-cell transcriptome analysis of >50,000 intratumoral immune cells after therapy treatment showed a gene signature suggestive of a change resulting from exposure to a mitogen, ligand, or antigen for which it is specific, as well as APC-to-T-cell adhesion. This transcriptional program also increased intratumoral Tcf1+ stem-like CD8+ T cells and altered the balance between terminally and progenitor-exhausted T cells favoring the latter. Overall, our data support the clinical investigation of this therapy in TNBC. SIGNIFICANCE: A combinatorial therapy in mouse models of breast cancer increases checkpoint inhibition by activating antigen-presenting cells, enhancing intratumoral Tcf1+ stem-like CD8+ T cells, and increasing progenitor exhausted CD8+ T cells.


Subject(s)
Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Cyclophosphamide/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Vinorelbine/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunity, Cellular , Mice , Mice, Inbred BALB C , Transcriptome , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology
7.
J Thromb Haemost ; 18(10): 2744-2750, 2020 10.
Article in English | MEDLINE | ID: mdl-32762140

ABSTRACT

BACKGROUND: During the course of COVID-19, the disease caused by the new coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), thrombotic phenomena and/or diffuse vascular damage are frequent, and viral elements have been observed within endothelial cells. OBJECTIVES: CD146 + circulating endothelial cells (CD146 + CECs) and their progenitors (CEPs) are increased in cardiovascular, thrombotic, infectious, and cancer diseases. The present study was designed to investigate their kinetics in novel coronavirus (COVID-19) patients. METHODS: We used a validated flow cytometry procedure to enumerate viable and apoptotic CD146 + CECs and CEPs in COVID-19 patients during the course of the disease and in patients who recovered. RESULTS: Viable CEPs per milliliter were significantly increased in COVID-19 patients compared with healthy controls. This increase was observed in patients with mild symptoms and not further augmented in patients with severe symptoms. In patients who recovered, CEPs decreased, but were in a range still significantly higher than normal controls. Regarding mature CD146 + CECs, in COVID-19 patients, their absolute number was similar to those observed in healthy controls, but the viable/apoptotic CD146 + CEC ratio was significantly different. Both mild and severe COVID-19 patients had significantly less apoptotic CD146 + CECs compared with healthy controls. Patients who recovered had significantly less CD146 + CECs per milliliter when compared with controls as well as to mild and severe COVID-19 patients. A positive correlation was found between the copies of SARS-CoV-2 RNA in the cellular fraction and apoptotic CEPs per milliliter in severe COVID-19 patients. CONCLUSIONS: CD146 + CECs and CEPs might be investigated as candidate biomarkers of endothelial damage in COVID-19 patients.


Subject(s)
Apoptosis , COVID-19 Nucleic Acid Testing , COVID-19/diagnosis , Endothelial Progenitor Cells/pathology , Flow Cytometry , Polymerase Chain Reaction , RNA, Viral/blood , SARS-CoV-2/genetics , Adult , Aged , Aged, 80 and over , Biomarkers/blood , CD146 Antigen/blood , COVID-19/blood , COVID-19/pathology , COVID-19/virology , Case-Control Studies , Endothelial Progenitor Cells/metabolism , Female , Host-Pathogen Interactions , Humans , Kinetics , Male , Middle Aged , Predictive Value of Tests , Severity of Illness Index , Viral Load
9.
Br J Cancer ; 118(10): 1329-1336, 2018 05.
Article in English | MEDLINE | ID: mdl-29695766

ABSTRACT

BACKGROUND: Anti-PD-1 and anti-PD-L1 checkpoint inhibitors (CIs) are clinically active in many types of cancer. However, only a minority of patients achieve a complete and/or long-lasting clinical response. We studied the effects of different doses of three widely used, orally active chemotherapeutics (vinorelbine, cyclophosphamide and 5-FU) over local and metastatic tumour growth, and the landscape of circulating and tumour-infiltrating immune cells involved in CI activity. METHODS: Immunocompetent Balb/c mice were used to generate models of breast cancer (BC) and B-cell lymphoma. Vinorelbine, cyclophosphamide and 5-FU (alone or in combination with CIs), were given at low-dose metronomic, medium, or maximum tolerable dosages. RESULTS: Cyclophosphamide increased circulating myeloid derived suppressor cells (MDSC). Vinorelbine, cyclophosphamide and 5-FU reduced circulating APCs. Vinorelbine and cyclophosphamide (at medium/high doses) reduced circulating Tregs. Cyclophosphamide (at low doses) and 5-FU (at medium doses) slightly increased circulating Tregs. Cyclophosphamide was the most potent drug in reducing circulating CD3+CD8+ and CD3+CD4+ T cells. Vinorelbine, cyclophosphamide and 5-FU reduced the number of circulating B cells, with cyclophosphamide showing the most potent effect. Vinorelbine reduced circulating NKs, whereas cyclophosphamide and 5-FU, at low doses, increased circulating NKs. In spite of reduced circulating T, B and NK effector cells, preclinical synergy was observed between chemotherapeutics and anti-PD-L1. Most-effective combinatorial regimens where associated with neoplastic lesions enriched in B cells, and, in BC-bearing mice (but not in mice with lymphoma) also in NK cells. CONCLUSIONS: Vinorelbine, cyclophosphamide and 5-FU have significant preclinical effects on circulating and tumour-infiltrating immune cells and can be used to obtain synergy with anti-PD-L1.


Subject(s)
B7-H1 Antigen/genetics , Breast Neoplasms/drug therapy , Lymphoma/drug therapy , Programmed Cell Death 1 Receptor/genetics , Animals , B7-H1 Antigen/immunology , B7-H1 Antigen/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Fluorouracil/administration & dosage , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/pathology , Mice , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/therapeutic use , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Vinorelbine/administration & dosage , Xenograft Model Antitumor Assays
10.
Cancer Res ; 77(18): 5169-5182, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28754674

ABSTRACT

A cell population with progenitor-like phenotype (CD45-CD34+) resident in human white adipose tissue (WAT) is known to promote the progression of local and metastatic breast cancer and angiogenesis. However, the molecular mechanisms of the interaction have not been elucidated. In this study, we identified two proteins that were significantly upregulated in WAT-derived progenitors after coculture with breast cancer: granulocyte macrophage colony-stimulating factor (GM-CSF) and matrix metallopeptidase 9 (MMP9). These proteins were released by WAT progenitors in xenograft and transgenic breast cancer models. GM-CSF was identified as an upstream modulator. Breast cancer-derived GM-CSF induced GM-CSF and MMP9 release from WAT progenitors, and GM-CSF knockdown in breast cancer cells neutralized the protumorigenic activity of WAT progenitors in preclinical models. GM-CSF neutralization in diet-induced obese mice significantly reduced immunosuppression, intratumor vascularization, and local and metastatic breast cancer progression. Similarly, MMP9 inhibition reduced neoplastic angiogenesis and significantly decreased local and metastatic tumor growth. Combined GM-CSF neutralization and MMP9 inhibition synergistically reduced angiogenesis and tumor progression. High-dose metformin inhibited GM-CSF and MMP9 release from WAT progenitors in in vitro and xenograft models. In obese syngeneic mice, metformin treatment mimicked the effects observed with GM-CSF neutralization and MMP9 inhibition, suggesting these proteins as new targets for metformin. These findings support the hypothesis that GM-CSF and MMP9 promote the protumorigenic effect of WAT progenitors on local and metastatic breast cancer. Cancer Res; 77(18); 5169-82. ©2017 AACR.


Subject(s)
Adipocytes/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Matrix Metalloproteinase 9/metabolism , Stem Cells/metabolism , Stromal Cells/pathology , Tumor Microenvironment/immunology , Adipocytes/pathology , Adult , Aged , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Stem Cells/pathology , Stromal Cells/immunology , Stromal Cells/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Sci Rep ; 6: 18673, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26728433

ABSTRACT

Metformin can induce breast cancer (BC) cell apoptosis and reduce BC local and metastatic growth in preclinical models. Since Metformin is frequently used along with Aspirin or beta-blockers, we investigated the effect of Metformin, Aspirin and the beta-blocker Atenolol in several BC models. In vitro, Aspirin synergized with Metformin in inducing apoptosis of triple negative and endocrine-sensitive BC cells, and in activating AMPK in BC and in white adipose tissue (WAT) progenitors known to cooperate to BC progression. Both Aspirin and Atenolol added to the inhibitory effect of Metformin against complex I of the respiratory chain. In both immune-deficient and immune-competent preclinical models, Atenolol increased Metformin activity against angiogenesis, local and metastatic growth of HER2+ and triple negative BC. Aspirin increased the activity of Metformin only in immune-competent HER2+ BC models. Both Aspirin and Atenolol, when added to Metformin, significantly reduced the endothelial cell component of tumor vessels, whereas pericytes were reduced by the addition of Atenolol but not by the addition of Aspirin. Our data indicate that the addition of Aspirin or of Atenolol to Metformin might be beneficial for BC control, and that this activity is likely due to effects on both BC and microenvironment cells.


Subject(s)
Antineoplastic Agents/pharmacology , Aspirin/pharmacology , Atenolol/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Metformin/pharmacology , Tumor Microenvironment/drug effects , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Apoptosis/drug effects , Biomarkers, Tumor , Breast Neoplasms/drug therapy , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Electron Transport Complex I/metabolism , Female , Humans , NAD/metabolism , Neoplasm Metastasis , Stem Cells/drug effects , Stem Cells/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Int J Cancer ; 136(6): E534-44, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25196138

ABSTRACT

The human white adipose tissue (WAT) contains progenitors with cooperative roles in breast cancer (BC) angiogenesis, local and metastatic progression. The biguanide Metformin (Met), commonly used for Type 2 diabetes, might have activity against BC and was found to inhibit angiogenesis in vivo. We studied Met and another biguanide, phenformin (Phe), in vitro and in vivo in BC models. In vitro, biguanides activated AMPK, inhibited Complex 1 of the respiratory chain and induced apoptosis of BC and WAT endothelial cells. In coculture, biguanides inhibited the production of several angiogenic proteins. In vivo, biguanides inhibited local and metastatic growth of triple negative and HER2+ BC in immune-competent and immune-deficient mice orthotopically injected with BC. Biguanides inhibited local and metastatic BC growth in a genetically engineered murine model model of HER2+ BC. In vivo, biguanides increased pimonidazole binding (but not HIF-1 expression) of WAT progenitors, reduced tumor microvessel density and altered the vascular pericyte/endothelial cell ratio, so that cancer vessels displayed a dysplastic phenotype. Phe was significantly more active than Met both in vitro and in vivo. Considering their safety profile, biguanides deserve to be further investigated for BC prevention in high-risk subjects, in combination with chemo and/or targeted therapy and/or as post-therapy consolidation or maintenance therapy for the prevention of BC recurrence.


Subject(s)
Breast Neoplasms/drug therapy , Metformin/pharmacology , Neovascularization, Pathologic/prevention & control , Phenformin/pharmacology , Tumor Microenvironment , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Line, Tumor , Electron Transport Complex I/antagonists & inhibitors , Female , Humans , Mice , Neoplasm Metastasis , Phosphorylation , TOR Serine-Threonine Kinases/metabolism
13.
PLoS One ; 9(12): e114713, 2014.
Article in English | MEDLINE | ID: mdl-25506915

ABSTRACT

BACKGROUND: The endothelium is not a homogeneous organ. Endothelial cell heterogeneity has been described at the level of cell morphology, function, gene expression, and antigen composition. As a consequence of the genetic, transcriptome and surrounding environment diversity, endothelial cells from different vascular beds have differentiated functions and phenotype. Detection of circulating endothelial cells (CECs) by flow cytometry is an approach widely used in cancer patients, and their number, viability and kinetic is a promising tool to stratify patient receiving anti-angiogenic treatment. METHODOLOGY/PRINCIPAL FINDINGS: Currently CECs are identified as positive for a nuclear binding antigen (DNA+), negative for the pan leukocyte marker CD45, and positive for CD31 and CD146. Following an approach recently validated in our laboratory, we investigated the expression of CD109 on CECs from the peripheral blood of healthy subject and cancer patients. The endothelial nature of these cells was validated by RT-PCR for the presence of m-RNA level of CDH5 (Ve-Cadherin) and CLDN5 (Claudin5), two endothelial specific transcripts. Before treatment, significantly higher levels of CD109+ CECs and viable CD109+CECs were found in breast cancer patients and glioblastoma patients compared to healthy controls, and their number significantly decreased after treatment. Higher levels of endothelial specific transcripts expressed in developing endothelial cells CLEC14a, TMEM204, ARHGEF15, GPR116, were observed in sorted CD109+CECs when compared to sorted CD146+CECs, suggesting that these genes can play an important role not only during embryogenesis but also in adult angiogenesis. Interestingly, mRNA levels of TEM8 (identified as Antrax Toxin Receptor1, Antrax1) were expressed in CD109+CECs+ but not in CD146+CECs. CONCLUSION: Taken together our results suggest that CD109 represent a rare population of circulating tumor endothelial cells, that play a potentially useful prognostic role in patients with glioblastoma. The role of CD109 expression in cancer vessel-specific endothelial cells deserves to be further investigated by gene expression studies.


Subject(s)
Antigens, CD/analysis , Endothelial Cells/pathology , Neoplasm Proteins/analysis , Neoplasms/blood , Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Adult , Aged , Antigens, CD/blood , Antigens, CD/genetics , Breast Neoplasms/blood , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CD146 Antigen/analysis , CD146 Antigen/blood , CD146 Antigen/genetics , Endothelial Cells/metabolism , Female , GPI-Linked Proteins/analysis , GPI-Linked Proteins/blood , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/blood , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Middle Aged , Neoplasm Proteins/blood , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplastic Cells, Circulating/metabolism
15.
PLoS One ; 8(9): e74345, 2013.
Article in English | MEDLINE | ID: mdl-24069296

ABSTRACT

BACKGROUND: Recent data suggest that circulating endothelial and progenitor cells (CECs and CEPs, respectively) may have predictive potential in cancer patients treated with bevacizumab, the antibody recognizing vascular endothelial growth factor (VEGF). Here we report on CECs and CEPs investigated in 68 patients affected by recurrent glioblastoma (rGBM) treated with bevacizumab and irinotecan and two Independent Datasets of rGBM patients respectively treated with bevacizumab alone (n=32, independent dataset A: IDA) and classical antiblastic chemotherapy (n=14, independent dataset B: IDB). METHODS: rGBM patients with KPS ≥50 were treated until progression, as defined by MRI with RANO criteria. CECs expressing CD109, a marker of tumor endothelial cells, as well as other CEC and CEP subtypes, were investigated by six-color flow cytometry. RESULTS: A baseline count of CD109+ CEC higher than 41.1/ml (1(st) quartile) was associated with increased progression free survival (PFS; 20 versus 9 weeks, P=0.008) and overall survival (OS; 32 versus 23 weeks, P=0.03). Longer PFS (25 versus 8 weeks, P=0.02) and OS (27 versus 17 weeks, P=0.03) were also confirmed in IDA with CD109+ CECs higher than 41.1/ml but not in IDB. Patients treated with bevacizumab with or without irinotecan that were free from MRI progression after two months of treatment had significant decrease of CD109+ CECs: median PFS was 19 weeks; median OS 29 weeks. The presence of two non-contiguous lesions (distant disease) at baseline was an independent predictor of shorter PFS and OS (P<0.001). CONCLUSIONS: Data encourage further studies on the predictive potential of CD109+ CECs in GBM patients treated with bevacizumab.


Subject(s)
Antigens, CD/metabolism , Endothelial Cells/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Neoplasm Proteins/metabolism , Adult , Aged , Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Disease Progression , Female , GPI-Linked Proteins/metabolism , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , Immunophenotyping , Irinotecan , Male , Middle Aged , Neoplasm Recurrence, Local , Prognosis , Treatment Outcome , Young Adult
16.
Cancer Res ; 73(19): 5880-91, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23918796

ABSTRACT

Obesity is associated with an increased frequency, morbidity, and mortality of several types of neoplastic diseases, including postmenopausal breast cancer. We found that human adipose tissue contains two populations of progenitors with cooperative roles in breast cancer. CD45(-)CD34(+)CD31(+)CD13(-)CCRL2(+) endothelial cells can generate mature endothelial cells and capillaries. Their cancer-promoting effect in the breast was limited in the absence of CD45(-)CD34(+)CD31(-)CD13(+)CD140b(+) mesenchymal progenitors/adipose stromal cells (ASC), which generated pericytes and were more efficient than endothelial cells in promoting local tumor growth. Both endothelial cells and ASCs induced epithelial-to-mesenchymal transition (EMT) gene expression in luminal breast cancer cells. Endothelial cells (but not ASCs) migrated to lymph nodes and to contralateral nascent breast cancer lesions where they generated new vessels. In vitro and in vivo, endothelial cells were more efficient than ASCs in promoting tumor migration and in inducing metastases. Granulocyte colony-stimulating factor (G-CSF) effectively mobilized endothelial cells (but not ASCs), and the addition of chemotherapy and/or of CXCR4 inhibitors did not increase endothelial cell or ASC blood mobilization. Our findings suggest that adipose tissue progenitor cells cooperate in driving progression and metastatic spread of breast cancer.


Subject(s)
Adipocytes/pathology , Adipose Tissue, White/pathology , Antigens, CD34/metabolism , Breast Neoplasms/pathology , Lung Neoplasms/secondary , Neovascularization, Pathologic/pathology , Stem Cells/pathology , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Female , Flow Cytometry , Fluorescent Antibody Technique , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/metabolism , Lymphatic Metastasis , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, CXCR4 , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Tumor Cells, Cultured
17.
Neoplasia ; 14(11): 1057-66, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23226099

ABSTRACT

Cell fusion plays a well-recognized physiological role during development, while its function during progression is still unclear. Here, we show that acute myeloid leukemia (AML) cells spontaneously fused with murine host cells in vivo. AML cells fused in most cases with mouse macrophages. Other targets of AML cell fusion were dendritic and endothelial cells. Cytogenetic and molecular analysis revealed that successive recipients conserved detectable amounts of parental DNA. Moreover, in a mouse AML1-ETO model where female AML1-ETO-leukemic cells, expressing CD45.2, were injected in congenic CD45.1 male mice AML cells, we found hybrid cells expressing both allelic types of CD45 and XXY set of sexual chromosomes. More importantly, the fusion protein AML1-ETO was transferred in the hybrid cells. When sorted hybrid cells were reinjected in a secondary recipient, they gave rise to leukemia with 100% penetrance and similar time of onset of leukemic cells. Our data indicate that in vivo fusion of cancer cells with host cells may be a mechanism of gene transfer for cancer dissemination and suggest that fused cells may be used to identify still unrecognized leukemogenic genes that are conserved in hybrid cells and able to perpetuate leukemia in vivo.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Macrophages/metabolism , Membrane Fusion , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD34/metabolism , CD47 Antigen/immunology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Dendritic Cells/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Female , Hematopoietic Stem Cells/metabolism , Humans , Hyaluronan Receptors/immunology , Hybrid Cells/metabolism , Leukemia, Lymphoid/genetics , Leukemia, Lymphoid/metabolism , Leukemia, Myeloid, Acute/genetics , Male , Membrane Fusion/drug effects , Mice , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RUNX1 Translocation Partner 1 Protein , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous
18.
Methods Mol Biol ; 904: 165-72, 2012.
Article in English | MEDLINE | ID: mdl-22890931

ABSTRACT

Results obtained from preclinical studies have shown that endothelial progenitor cells (EPCs) play a crucial role in tumor growth and metastasis. In the clinic, EPCs are present in the peripheral blood of cancer patients in higher numbers than in healthy subjects. These cells are mobilized from the bone marrow compartment to the periphery in response to certain cytokines and growth factors. Growing body of evidence suggests that following acute cytotoxic drug therapy levels of circulating EPCs (CEPs) can change significantly in both mouse and human. These changes may predict the efficacy of some anticancer drug treatments. Therefore, the validation and standardization of a procedure to detect CEPs and monitor their kinetic is an important step towards the use of such cells as a possible biomarker to predict clinical outcome. In this chapter, we describe a flow cytometry technique to detect CEPs obtained from human blood specimens stored in both fresh and frozen conditions.


Subject(s)
Endothelial Cells/metabolism , Neoplasms/metabolism , Stem Cells/metabolism , Antigens, CD/metabolism , Flow Cytometry , Humans , Immunophenotyping , Leukocytes, Mononuclear/metabolism , Neovascularization, Pathologic/metabolism
19.
Exp Diabetes Res ; 2012: 274363, 2012.
Article in English | MEDLINE | ID: mdl-22474419

ABSTRACT

Chronic diabetic complications result from an imbalance between vascular damage and regeneration. Several circulating lineage-committed progenitor cells have been implicated, but no data are available on pericyte progenitor cells (PPCs). Based on the evidence that PPCs increase in cancer patients after chemotherapy, we explored whether circulating PPC levels are affected by glucose control in type 2 diabetic patients, in relation to the presence of chronic complications. We enumerated peripheral blood PPCs as Syto16+CD45-CD31-CD140b+ events by flow cytometry at baseline and after 3 and 6 months of glucose control by means of add-on basal insulin therapy on top of oral agents in 38 poorly controlled type 2 diabetic patients. We found that, in patients with microangiopathy (n = 23), the level of circulating PPCs increased about 2 fold after 3 months and then returned to baseline at 6 months. In patients without microangiopathy (control group, n = 15), PPCs remained fairly stable during the whole study period. No relationship was found between change in PPCs and macroangiopathy (either peripheral, coronary, or cerebrovascular). We conclude that glucose control transiently mobilizes PPCs diabetic patients with microangiopathy. Increase in PPCs may represent a vasoregenerative event or may be a consequence of ameliorated glucose control on microvascular lesions.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/drug therapy , Hypoglycemic Agents/pharmacology , Insulin, Long-Acting/pharmacology , Pericytes/drug effects , Aged , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetic Angiopathies/blood , Flow Cytometry , Humans , Hypoglycemic Agents/therapeutic use , Insulin Detemir , Insulin Glargine , Insulin, Long-Acting/therapeutic use , Middle Aged , Stem Cells/drug effects , Treatment Outcome
20.
Recent Results Cancer Res ; 195: 163-70, 2012.
Article in English | MEDLINE | ID: mdl-22527503

ABSTRACT

The roles of circulating endothelial cells (CECs) and circulating endothelial progenitors (CEPs) are currently being investigated in several diseases including cancer and metastases development. Preclinical and clinical data suggest that CEC enumeration might be useful to identify patients who might benefit from anti-angiogenic treatments while CEPs seem to have a "catalytic" role in different steps of cancer progression and recurrence after therapy. The definition of CEC and CEP phenotypes and the standardization of CEC and CEP enumeration procedures are highly warranted to use these cells as biomarkers in clinical trials in oncology, and to compare results from different studies.


Subject(s)
Endothelial Cells/pathology , Neoplasms/blood , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Stem Cells/pathology , Animals , Humans , Neoplasm Metastasis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...