Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Biomedicines ; 11(2)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36830940

ABSTRACT

The two most developed biomarkers in liquid biopsy (LB)-circulating tumor cells and circulating tumor DNA-have been joined by the analysis of extracellular vesicles (EVs). EVs are lipid-bilayer enclosed structures released by all cell types containing a variety of molecules, including DNA, mRNA and miRNA. However, fast, efficient and a high degree of purity isolation technologies are necessary for their clinical routine implementation. In this work, the use of ExoGAG, a new easy-to-use EV isolation technology, was validated for the isolation of EVs from plasma and urine samples. After demonstrating its efficiency, an analysis of the genetic material contained in the EVs was carried out. Firstly, the sensitivity of the detection of point mutations in DNA from plasma EVs isolated by ExoGAG was analyzed. Then, a pilot study of mRNA expression using the nCounter NanoString platform in EV-mRNA from a healthy donor, a benign prostate hyperplasia patient and metastatic prostate cancer patient plasma and urine samples was performed, identifying the prostate cancer pathway as one of the main ones. This work provides evidence for the value of using ExoGAG for the isolation of EVs from plasma and urine samples, enabling downstream applications of the analysis of their genetic cargo.

2.
Sci Rep ; 11(1): 23231, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34853364

ABSTRACT

Homing of circulating tumour cells (CTC) at distant sites represents a critical event in metastasis dissemination. In addition to physical entrapment, probably responsible of the majority of the homing events, the vascular system provides with geometrical factors that govern the flow biomechanics and impact on the fate of the CTC. Here we mathematically explored the distribution of velocities and the corresponding streamlines at the bifurcations of large blood vessel and characterized an area of low-velocity at the carina of bifurcation that favours the residence of CTC. In addition to this fluid physics effect, the adhesive capabilities of the CTC provide with a biological competitive advantage resulting in a marginal but systematic arrest as evidenced by dynamic in vitro recirculation in Y-microchannels and by perfusion in in vivo mice models. Our results also demonstrate that viscosity, as a main determinant of the Reynolds number that define flow biomechanics, may be modulated to limit or impair CTC accumulation at the bifurcation of blood vessels, in agreement with the apparent positive effect observed in the clinical setting by anticoagulants in advanced oncology disease.


Subject(s)
Blood Flow Velocity , Hemodynamics , Neoplastic Cells, Circulating , Animals , Cell Adhesion , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells , Humans , Leukocytes, Mononuclear , Mice , Models, Cardiovascular , Models, Theoretical
3.
Biomed Pharmacother ; 140: 111744, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34049223

ABSTRACT

Endometrial cancer (EC) is the most common neoplasm of the female reproductive tract in the developed world. Patients usually are diagnosed in early stage having a good prognosis. However, up to 20-25% of patients are diagnosed in advanced stages and have a higher risk of recurrence, making the prognosis worse. Previously studies identified ANXA2 as a predictor of recurrent disease in EC even in low risk patients. Furthermore, Circulating Tumor Cells (CTC) released from the primary tumor into the bloodstream, are plasticity entities responsible of the process of metastasis, becoming into an attractive clinical target. In this work we validated ANXA2 expression in CTC from high-risk EC patients. After that, we modelled in vitro and in vivo the tumor cell attachment of ANXA2-expressing CTC to the endothelium and the homing for the generation of micrometastasis. ANXA2 overexpression does not provide an advantage in the adhesion process of CTC, but it could be playing an important role in more advanced steps, conferring a greater homing capacity. We also performed a high-throughput screening (HTS) for compounds specifically targeting ANXA2, and selected Daunorubicin as candidate hit. Finally, we validated Daunorubicin in a 3D transendothelial migration system and also in a in vivo model of advanced EC, demonstrating the ability of Daunorubicin to inhibit the proliferation of ANXA2-overexpressing tumor cells.


Subject(s)
Annexin A2/metabolism , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Animals , Annexin A2/genetics , Cell Adhesion , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Daunorubicin/pharmacology , Daunorubicin/therapeutic use , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endothelium/physiology , Female , High-Throughput Screening Assays , Humans , Liquid Biopsy , Mice , Models, Biological , Neoplastic Cells, Circulating
4.
Gynecol Oncol ; 161(3): 681-686, 2021 06.
Article in English | MEDLINE | ID: mdl-33795131

ABSTRACT

OBJECTIVE: Despite radical surgery and chemotherapy, most patients with ovarian cancer die due to disease progression. M-Trap is an implantable medical device designed to capture peritoneal disseminated tumor cells with the aim to focalize the disease. This trial analyzed the safety and performance of the device. METHODS: This first-in-human prospective, multi-center, non-blinded, single-arm study enrolled 23 women with high-grade serous advanced ovarian cancer. After primary or interval debulking surgery, 3 M-Trap devices were placed in the peritoneum of the abdominal cavity. 18-months post-implantation or at disease progression, devices were initially removed by laparoscopy. The primary safety endpoint was freedom from device and procedure-related major adverse events (MAEs) through 6-months post-implantation compared to an historical control. The primary performance endpoint was histopathologic evidence of tumor cells capture. RESULTS: Only one major adverse event was attributable to the device. 18 women were free of device and procedure related MAEs (78.3%). However, the primary safety endpoint was not achieved (p = 0.131), primarily attributable to the greater surgical complexity of the M-Trap patient population. 62% of recurrent patients demonstrated tumor cell capture in at least one device with a minimal tumor cell infiltration. No other long-term device-related adverse events were reported. The secondary performance endpoint demonstrated a lack of disease focalization. CONCLUSIONS: The M-Trap technology failed to meet its primary safety objective, although when adjusted for surgical complexity, the study approved it. Likewise, the devices did not demonstrate the anticipated benefits in terms of tumor cell capture and disease focalization in recurrent ovarian cancer.


Subject(s)
Carcinoma, Ovarian Epithelial/surgery , Cytoreduction Surgical Procedures/instrumentation , Neoplasm Recurrence, Local/surgery , Ovarian Neoplasms/surgery , Peritoneal Neoplasms/surgery , Adult , Aged , Carcinoma, Ovarian Epithelial/secondary , Female , Humans , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local/pathology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Prospective Studies , Spain , Treatment Outcome
5.
Dis Model Mech ; 13(6)2020 06 17.
Article in English | MEDLINE | ID: mdl-32764154

ABSTRACT

Metastasis is facilitated by the formation of pre-metastatic niches through the remodelling of the extracellular matrix (ECM) promoted by haematopoietic and stromal cells. The impact of these primed sites is pronounced for intraperitoneal metastases, where the cavity-exposed ECM supports the attachment of the disseminating tumour cells. Likewise, implantation of biomaterial scaffolds influences metastatic progression systemically through a foreign body reaction (FBR). In this study, we integrated the concept of creating an artificial niche to capture tumour cells actively disseminating in the peritoneal cavity with a therapeutic strategy modulating the interactions of metastatic cells with the ECM. The aim was to transform a disseminated disease into a focal disease. For this, we designed and developed a 'biomimetic' ECM composed of a nonresorbable three-dimensional scaffold with collagen coating and characterized the FBR to the implanted biomaterial. We also analysed the safety of the implanted devices and their ability to capture tumour cells in different murine preclinical models of advanced ovarian cancer. Implantation of the biomimetic devices resulted in an initial inflammatory reaction that transformed progressively into a fibrous connective tissue response. The adhesive capabilities of the scaffold were improved with the ancillary effect of the FBR and showed clinical utility in terms of the efficacy of capture of tumour cells, disease focalization and survival benefit. These results demonstrated the performance and safety of this 'biomimetic' ECM in preclinical models of advanced ovarian cancer. Translated into the clinical setting, this new therapeutic strategy represents the possibility for control of peritoneal carcinomatosis upon primary ovarian debulking surgery and to expand the percentage of patients who are candidates for second rescue surgeries at the time of relapse.


Subject(s)
Biomimetic Materials , Biomimetics/instrumentation , Cell Adhesion , Extracellular Matrix/pathology , Foreign-Body Migration/pathology , Ovarian Neoplasms/therapy , Peritoneal Neoplasms/prevention & control , Tissue Scaffolds , Animals , Cell Line, Tumor , Extracellular Matrix/metabolism , Female , Foreign-Body Migration/metabolism , Humans , Mice, SCID , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Time Factors , Tumor Microenvironment , Xenograft Model Antitumor Assays
6.
Cells ; 9(5)2020 05 14.
Article in English | MEDLINE | ID: mdl-32423054

ABSTRACT

BACKGROUND: Recent studies showed a relevant role of hematogenous spread in ovarian cancer and the interest of circulating tumor cells (CTCs) monitoring as a prognosis marker. The aim of the present study was the characterization of CTCs from ovarian cancer patients, paying special attention to cell plasticity characteristics to better understand the biology of these cells. METHODS: CTCs isolation was carried out in 38 patients with advanced high-grade serous ovarian cancer using in parallel CellSearch and an alternative EpCAM-based immunoisolation followed by RT-qPCR analysis to characterize these cells. RESULTS: Epithelial CTCs were found in 21% of patients, being their presence higher in patients with extraperitoneal metastasis. Importantly, this population was characterized by the expression of epithelial markers as MUC1 and CK19, but also by genes associated with mesenchymal and more malignant features as TIMP1, CXCR4 and the stem markers CD24 and CD44. In addition, we evidenced the relevance of TIMP1 expression to promote tumor proliferation, suggesting its interest as a therapeutic target. CONCLUSIONS: Overall, we evidenced the utility of the molecular characterization of EpCAM+ CTCs from advanced ovarian cancer patients to identify biomarkers with potential applicability for disseminated disease detection and as therapeutic targets such as TIMP1.


Subject(s)
Molecular Targeted Therapy , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Plasticity , Cell Proliferation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Middle Aged , Neoplasm Invasiveness , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Xenograft Model Antitumor Assays , Young Adult , Zebrafish
7.
J Clin Med ; 9(2)2020 Feb 21.
Article in English | MEDLINE | ID: mdl-32098121

ABSTRACT

The incidence and mortality of endometrial cancer (EC) have risen in recent years, hence more precise management is needed. Therefore, we combined different types of liquid biopsies to better characterize the genetic landscape of EC in a non-invasive and dynamic manner. Uterine aspirates (UAs) from 60 patients with EC were obtained during surgery and analyzed by next-generation sequencing (NGS). Blood samples, collected at surgery, were used for cell-free DNA (cfDNA) and circulating tumor cell (CTC) analyses. Finally, personalized therapies were tested in patient-derived xenografts (PDXs) generated from the UAs. NGS analyses revealed the presence of genetic alterations in 93% of the tumors. Circulating tumor DNA (ctDNA) was present in 41.2% of cases, mainly in patients with high-risk tumors, thus indicating a clear association with a more aggressive disease. Accordingly, the results obtained during the post-surgery follow-up indicated the presence of ctDNA in three patients with progressive disease. Moreover, 38.9% of patients were positive for CTCs at surgery. Finally, the efficacy of targeted therapies based on the UA-specific mutational landscape was demonstrated in PDX models. Our study indicates the potential clinical applicability of a personalized strategy based on a combination of different liquid biopsies to characterize and monitor tumor evolution, and to identify targeted therapies.

8.
J Proteome Res ; 18(3): 1043-1053, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30585730

ABSTRACT

Endometrial cancer (EC) is the most frequent gynecological cancer. Tumor dissemination affecting ∼20% of EC patients is characterized at the primary carcinoma by epithelial-to-mesenchymal transition (EMT) associated with myometrial infiltration. At distant sites, the interaction of circulating tumor cells (CTCs) with the microenvironment is crucial for metastatic colonization, with a participation of the extracellular vesicles (EVs). We comprehensively approached these primary and secondary sites to study the impact of tumor EVs on the metastatic efficiency of CTCs in EC. Tumor EVs in circulation reproduce the epithelial phenotype predominant in the primary carcinoma, whereas CTCs are characterized by an EMT phenotype. We modeled this EMT-related clinical scenario in the Hec1A endometrial cell line and characterized the epithelial-like EVs in circulation by SILAC proteome analysis. The identification of proteins involved in cell-cell and cell-matrix interaction and binding, together with in vitro evidence of an improved adhesion of CTC to a functionalized endothelium, suggests a contribution of the epithelial-like EVs in the homing of CTCs at metastatic sites. Accordingly, adhesion protein LGALS3BP was found to be significantly enriched in circulating EVs from a cohort of EC patients with a high risk of recurrence by targeted proteomics (multiple reaction monitoring), highlighting its potential in liquid biopsy in EC.


Subject(s)
Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Endometrial Neoplasms/genetics , Proteome/genetics , Proteomics , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/blood , Biomarkers, Tumor/blood , Endometrial Neoplasms/blood , Endometrial Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Extracellular Vesicles/genetics , Extracellular Vesicles/pathology , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Isotope Labeling , Middle Aged , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Microenvironment/genetics
9.
Oncotarget ; 9(24): 16648-16664, 2018 Mar 30.
Article in English | MEDLINE | ID: mdl-29682175

ABSTRACT

Endometrial cancer (EC) is the sixth deadliest cancer in women. The depth of myometrial invasion is one of the most important prognostic factors, being directly associated with tumor recurrence and mortality. In this study, ALCAM, a previously described marker of EC recurrence, was studied by immunohistochemistry at the superficial and the invasive tumor areas from 116 EC patients with different degree of myometrial invasion and related to a set of relevant epithelial and mesenchymal markers. ALCAM expression presented a heterogeneous functionality depending on its localization, it correlated with epithelial markers (E-cadherin/ß-catenin) at the superficial area, and with mesenchymal markers at the invasive front (COX-2, SNAIL, ETV5, and MMP-9). At the invasive front, ALCAM-negativity was an independent marker of myometrial invasion. This negativity, together with an increase of soluble ALCAM in uterine aspirates from patients with an invasive EC, and its positive correlation with MMP-9 levels, suggested that ALCAM shedding by MMP-9 occurs at the invasive front. In vivo and in vitro models of invasive EC were generated by ETV5-overexpression. In those, we demonstrated that ALCAM shedding was related to a more invasive pattern and that full-ALCAM recovery reverted most of the ETV5-cells mesenchymal abilities, partially through a p-ERK dependent-manner.

10.
Int J Mol Sci ; 18(6)2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28608814

ABSTRACT

Colorectal cancer (CRC) is one of the major causes of cancer-related deaths. Early detection of tumor relapse is crucial for determining the most appropriate therapeutic management. In clinical practice, computed tomography (CT) is routinely used, but small tumor changes are difficult to visualize, and reliable blood-based prognostic and monitoring biomarkers are urgently needed. The aim of this study was to prospectively validate a gene expression panel (composed of GAPDH, VIL1, CLU, TIMP1, TLN1, LOXL3 and ZEB2) for detecting circulating tumor cells (CTCs) as prognostic and predictive tool in blood samples from 94 metastatic CRC (mCRC) patients. Patients with higher gene panel expression before treatment had a reduced progression-free survival (PFS) and overall-survival (OS) rates compared with patients with low expression (p = 0.003 and p ≤ 0.001, respectively). Patients with increased expression of CTCs markers during treatment presented PFS and OS times of 8.95 and 11.74 months, respectively, compared with 14.41 and 24.7 for patients presenting decreased expression (PFS; p = 0.020; OS; p ≤ 0.001). Patients classified as non-responders by CTCs with treatment, but classified as responders by CT scan, showed significantly shorter survival times (PFS: 8.53 vs. 11.70; OS: 10.37 vs. 24.13; months). In conclusion, our CTCs detection panel demonstrated efficacy for early treatment response assessment in mCRC patients, and with increased reliability compared to CT scan.


Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplastic Cells, Circulating/pathology , Biomarkers, Tumor/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Neoplastic Cells, Circulating/metabolism , Prognosis , Prospective Studies , Treatment Outcome
11.
Cancer Res ; 77(13): 3431-3441, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28536280

ABSTRACT

The interaction between circulating tumor cells (CTC) and endothelial cells during extravasation is a critical process during metastatic colonization, but its mechanisms remain poorly characterized. Here we report that the luminal side of liver blood vessels contains fibronectin deposits that are enriched in mice bearing primary tumors and are also present in vessels from human livers affected with metastases. Cancer cells attached to endothelial fibronectin deposits via talin1, a major component of focal adhesions. Talin1 depletion impaired cancer cell adhesion to the endothelium and transendothelial migration, resulting in reduced liver metastasis formation in vivo Talin1 expression levels in patient CTC's correlated with prognosis and therapy response. Together, our findings uncover a new mechanism for liver metastasis formation involving an active contribution of hepatic vascular fibronectin and talin1 in cancer cells. Cancer Res; 77(13); 3431-41. ©2017 AACR.


Subject(s)
Fibronectins/metabolism , Liver Neoplasms/blood , Liver Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Animals , Cell Adhesion/physiology , Cell Line, Tumor , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Transendothelial and Transepithelial Migration
12.
J Natl Cancer Inst ; 107(9)2015 Sep.
Article in English | MEDLINE | ID: mdl-26150590

ABSTRACT

BACKGROUND: Remodeling targeted tissues for reception of tumor cells metastasizing from primary lesions is a consequence of communication between the tumor and the environment that governs metastasis. This study describes a novel approach that aims to disrupt the process of metastasis by interfering with this intense dialogue. METHODS: Proteomics and adhesion assays identified exosomes purified from the ascitic fluid of ovarian cancer patients (n = 9) as intermediaries of tumor cell attachment. A novel tumor cell capture device was fabricated by embedding exosomes onto a 3D scaffold (metastatic trap [M-Trap]). Murine models of ovarian metastasis (n = 3 to 34 mice per group) were used to demonstrate the efficacy of M-Trap to capture metastatic cells disseminating in the peritoneal cavity. Kaplan-Meier survival curves were used to estimate cumulative survival probabilities. All statistical tests were two-sided. RESULTS: The exosome-based M-Trap device promoted tumor cell adhesion with a nonpharmacological mode of action. M-Trap served as a preferential site for metastasis formation and completely remodeled the pattern of peritoneal metastasis in clinically relevant models of ovarian cancer. Most importantly, M-Trap demonstrated a statistically significant benefit in survival outcomes, with mean survival increasing from 117.5 to 198.8 days in the presence of M-Trap; removal of the device upon tumor cell capture further improved survival to a mean of 309.4 days (P < .001). CONCLUSIONS: A potent artificial premetastatic niche based on exosomes is an effective approach to impair the crosstalk between metastatic cells and their environment. In the clinical setting, the capacity to modulate the pattern of dissemination represents an opportunity to control the process of metastasis. In summary, M-Trap transforms a systemic, fatal disease into a focalized disease where proven therapeutic approaches such as surgery can extend survival.


Subject(s)
Exosomes , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Animals , Ascitic Fluid/pathology , Cell Adhesion , Cell Line, Tumor , Female , Humans , Kaplan-Meier Estimate , Mice , Neoplasms, Experimental
13.
Clin Exp Metastasis ; 32(5): 467-78, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25924802

ABSTRACT

Endometrial cancer is the most frequent malignancy of the female genital tract in western countries. Our group has previously characterized the upregulation of the transcription factor ETV5 in endometrial cancer with a specific and significant increase in those tumor stages associated with myometrial invasion. We have shown that ETV5 overexpression in Hec1A endometrial cancer cells induces epithelial to mesenchymal transition resulting in the acquisition of migratory and invasive capabilities. In the present work, we have identified Nidogen 1 (NID1) and Nuclear Protein 1 (NUPR1) as direct transcriptional targets of ETV5 in endometrial cancer cells. Inhibition of NID1 and NUPR1 in ETV5 overexpressing cells reduced cell migration and invasion in vitro and reduced tumor growth and dissemination in an orthotopic endometrial cancer model. Importantly, we confirmed a significant increase of NUPR1 and NID1 protein expression in the invasion front of the tumor compared to their paired superficial zone, concomitant to ETV5 overexpression. Altogether, we conclude that NID1 and NUPR1 are novel targets of ETV5 and are actively cooperating with ETV5 at the invasion front of the tumor in the acquisition of an invasive phenotype to jointly drive endometrial cancer invasion.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Movement , Cell Proliferation , DNA-Binding Proteins/metabolism , Endometrial Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Transcription Factors/metabolism , Animals , Apoptosis , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Female , Humans , Immunoenzyme Techniques , Luciferases/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Int J Cancer ; 136(8): 1863-73, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25219463

ABSTRACT

Endometrial carcinomas, the most common malignant tumour of the female genital tract, are usually diagnosed at an early stage with uterine-confined disease and an overall favourable prognosis. However, up to 20% of endometrial carcinomas will end up in recurrent disease, associated with a drop in survival and representing the major clinical challenge. Management of this group of risk patients relies on robust biomarkers that may predict which endometrial carcinomas will relapse. For this, we performed a proteomic analysis comparing primary lesions with recurrences and identified ANXA2 as a potential biomarker associated with recurrent disease that we further validated in an independent series of samples by immunohistochemistry. We demonstrated in vitro a role for ANXA2 in the promotion of metastasis rather than interfering with sensitivity to radio/chemotherapy. In addition, ANXA2 silencing resulted in a reduced metastatic pattern in a mice model of endometrial cancer dissemination, with a limited presence of circulating tumor cells. Finally, a retrospective study in a cohort of 93 patients showed that ANXA2 effectively predicted those endometrioid endometrial carcinomas that finally recurred. Importantly, ANXA2 demonstrated a predictive value also among low risk Stage I endometrioid endometrial carcinomas, highlighting the clinical utility of ANXA2 biomarker as predictor of recurrent disease in endometrial cancer. Retrospective and prospective studies are ongoing to validate ANXA2 as a potential tool for optimal stratification of patients susceptible to receive radical surgery and radio/chemotherapy.


Subject(s)
Annexin A2/blood , Biomarkers, Tumor/blood , Endometrial Neoplasms/blood , Endometrial Neoplasms/diagnosis , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/diagnosis , Aged , Animals , Female , Humans , Mice , Mice, Nude , Neoplasm Metastasis/diagnosis , Neoplastic Cells, Circulating/metabolism , Proteomics/methods , Retrospective Studies
15.
Mol Cancer ; 13: 223, 2014 Sep 27.
Article in English | MEDLINE | ID: mdl-25261936

ABSTRACT

BACKGROUND: About 20% of patients diagnosed with endometrial cancer (EC) are considered high-risk with unfavorable prognosis. In the framework of the European Network for Individualized Treatment in EC (ENITEC), we investigated the presence and phenotypic features of Circulating Tumor Cells (CTC) in high-risk EC patients. METHODS: CTC isolation was carried out in peripheral blood samples from 34 patients, ranging from Grade 3 Stage IB to Stage IV carcinomas and recurrences, and 27 healthy controls using two methodologies. Samples were subjected to EpCAM-based immunoisolation using the CELLection™ Epithelial Enrich kit (Invitrogen, Dynal) followed by RTqPCR analysis. The phenotypic determinants of endometrial CTC in terms of pathogenesis, hormone receptor pathways, stem cell markers and epithelial to mesenchymal transition (EMT) drivers were asked. Kruskal-Wallis analysis followed by Dunn's post-test was used for comparisons between groups. Statistical significance was set at p < 0.05. RESULTS: EpCAM-based immunoisolation positively detected CTC in high-risk endometrial cancer patients. CTC characterization indicated a remarkable plasticity phenotype defined by the expression of the EMT markers ETV5, NOTCH1, SNAI1, TGFB1, ZEB1 and ZEB2. In addition, the expression of ALDH and CD44 pointed to an association with stemness, while the expression of CTNNB1, STS, GDF15, RELA, RUNX1, BRAF and PIK3CA suggested potential therapeutic targets. We further recapitulated the EMT phenotype found in endometrial CTC through the up-regulation of ETV5 in an EC cell line, and validated in an animal model of systemic dissemination the propensity of these CTC in the accomplishment of metastasis. CONCLUSIONS: Our results associate the presence of CTC with high-risk EC. Gene-expression profiling characterized a CTC-plasticity phenotype with stemness and EMT features. We finally recapitulated this CTC-phenotype by over-expressing ETV5 in the EC cell line Hec1A and demonstrated an advantage in the promotion of metastasis in an in vivo mouse model of CTC dissemination and homing.


Subject(s)
Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Gene Expression Profiling , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Aged , Animals , Cell Separation , DNA-Binding Proteins/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , Neoplasm Metastasis , Phenotype , Risk Factors , Transcription Factors/metabolism
16.
Carcinogenesis ; 35(12): 2679-86, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25233929

ABSTRACT

Myometrial infiltration represents a main clinical determinant of endometrial carcinomas (EC) presenting as aggressive high-grade deeply invasive neoplasms, substantially associated with risk of recurrence and death. The up-regulation of ETV5 transcription factor linked to the promotion of epithelial to mesenchymal transition is considered as a basic mechanism underlying the initial steps of EC invasion. In this work, we aimed to investigate the transcription program of tumor invasion regulated by ETV5. We performed a comparative Chip-on-chip analysis at invasive front and superficial area of human EC. ETV5 specific binding to promoter regions of genes related to cellular migration, adhesion and invasion at deep invasion tumor areas highlighted the relevance of neural networks associated with cellular plasticity. Interestingly, brain-derived neurotrophic factor (BDNF) demonstrated a principal role orchestrating ETV5-mediated epithelial-to-mesenchymal transition in endometrial cancer. Impairment of the BDNF/tropomyosin-related kinase B (TrkB)/extracellular signal-regulated kinase axis in endometrial cancer cell lines reversed the aggressive and invasive phenotype promoted by the up-regulation of ETV5 at the invasive front of EC. Likewise, BDNF directly impacted on the efficiency of ETV5 promoted metastasis in a mice model of endometrial distant dissemination. These results translate the recognized role of BDNF/TrkB on neural plasticity into a relevant cancer metastasis event; suggest common mechanisms shared by neural development and tumor invasion; and offer new therapeutic opportunities specifically directed against disseminated disease in endometrial cancer.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell Movement , DNA-Binding Proteins/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Transcription Factors/genetics , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/genetics , Cell Proliferation , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Endometrial Neoplasms/genetics , Female , Fluorescent Antibody Technique , Humans , Mice , Neoplasm Invasiveness , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Tumor Cells, Cultured
17.
Anticancer Res ; 33(12): 5295-300, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24324062

ABSTRACT

BACKGROUND: Zoledronic acid effectively reduces skeletal events in patients with metastatic disease. The results of pre-clinical and emerging clinical data suggest an additional activity of zoledronic acid as an antitumor agent, interfering with the growth and dissemination of malignant cells. However, the mechanisms by which zoledronic acid impairs tumor progression are practically unknown. In the present study, we aimed to investigate the impact of zoledronic acid on invasion and colony formation ability of different human tumour cell lines. MATERIALS AND METHODS: Human ovarian (SKOV3), colonic (HCT116), endometrial (HEC1A and Ishikawa) and breast cancer (MCF-7, MDA-MB-231, HCC1937, SKBR3 and T47D) cell lines were treated with different concentrations (10-100 µM) of zoledronic acid and analyzed using 3D assays to test their invasiveness and their ability to grow anchorage-independently, both hallmarks of aggressive tumor cell behavior. RESULTS: The most intense effect of the drug on tumor invasion was observed on MDA-MB-231 cells, but at high concentrations HEC1A, SKOV3 and SKBR3 cells also exhibited reduced invasion capacity. We also found a significant reduction of colony formation under zoledronic acid treatment in MCF-7, T47-D, HCT116, Ishikawa, HEC1A and SKOV3 cells. CONCLUSION: Zoledronic acid presents an interesting potential for use as anti-metastatic agent for different solid tumor types, affecting relevant steps of tumor dissemination.


Subject(s)
Diphosphonates/pharmacology , Imidazoles/pharmacology , Neoplasm Metastasis/prevention & control , Cell Line, Tumor , Humans , Neoplasm Invasiveness/prevention & control , Zoledronic Acid
18.
PLoS One ; 7(7): e40476, 2012.
Article in English | MEDLINE | ID: mdl-22811761

ABSTRACT

Metastatic colorectal cancer (mCRC) relies on the detachment of aggressive malignant cells from the primary tumor into the bloodstream and, concordantly, the presence of these Circulating Tumor Cells (CTC) is associated with a poor prognosis. In this work, the molecular characterization of CTC from mCRC patients was approached, with the aim of understanding their biology and improving their clinical utility in the management of colorectal cancer patients. For this, EpCAM-based immunoisolation of CTC was combined with whole transcriptome amplification and hybridization onto cDNA microarrays. Gene expression data from mCRC patients, once the background of unspecific immunoisolation from a group of controls had been subtracted, resulted in 410 genes that characterized the CTC population. Bioinformatics were used for the biological interpretation of the data, revealing that CTC are characterized by genes related to cell movement and adhesion, cell death and proliferation, and cell signalling and interaction. RTqPCR on an independent series of mCRC patients and controls was used for the validation of a number of genes related to the main cellular functions characterizing the CTC population. Comparison between primary carcinomas and lung and liver metastases further involved the CTC-genes in the promotion of metastasis. Moreover, the correlation of CTC-gene expression with clinical parameters demonstrated detection and prognosis significance. In conclusion, the molecular characterization of CTC from mCRC patients and the identification of diagnostic and prognostic biomarkers represent an innovative and promising approach in the clinical management of this type of patients.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Colorectal Neoplasms/diagnosis , Humans , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Oligonucleotide Array Sequence Analysis , Prognosis , Transcriptome
19.
J Cell Mol Med ; 16(10): 2342-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22304365

ABSTRACT

The accuracy in the diagnosis of metastatic colorectal cancer (mCRC) represents one of the challenges in the clinical management of patients. The detection of circulating tumour cells (CTC) is becoming a promising alternative to current detection techniques, as it focuses on one of the players of the metastatic disease and it should provide with more specific and sensitive detection rates. Here, we describe an improved method of detection of CTC from mCRC patients by combining immune-enrichment, optimal purification of RNA from very low cell numbers, and the selection of accurate PCR probes. As a result, we obtained a logistic model that combines GAPDH and VIL1 normalized to CD45 rendering powerful results in the detection of CTC from mCRC patients (AUROC value 0.8599). We further demonstrated the utility of this model at the clinical setting, as a reliable prognosis tool to determine progression-free survival in mCRC patients. Overall, we developed a strategy that ameliorates the specificity and sensitivity in the detection of CTC, resulting in a robust and promising logistic model for the clinical management of metastatic colorectal cancer patients.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Epithelial Cells/metabolism , Female , Humans , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Logistic Models , Male , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Middle Aged , Prognosis , Reproducibility of Results , Sensitivity and Specificity
20.
Mol Cancer Ther ; 10(8): 1357-66, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21613448

ABSTRACT

Endometrial cancer is among the three most common cancers in females in industrialized countries. In the majority of cases, the tumor is confined to the uterus at the time of diagnosis and presents a good prognosis. However, after primary surgery, 15% to 20% of these tumors recur and have limited response to systemic therapy. We carried out gene expression profiling of high-risk recurrence endometrial cancers to identify new therapeutic approaches targeting the molecular pathways involved in the acquisition of an aggressive tumor phenotype. A microarray gene-expression analysis on a total of 51 human endometrial carcinomas revealed 77 genes specifically altered in high-risk recurrence tumors (P < 0.001). The bioinformatics analysis of gene-gene interactions and molecular relationships among these genes pointed to a prominent role for TGF-ß1 signaling in the acquisition of an aggressive phenotype. We further showed that TGF-ß1 has a principal role at the initiation of endometrial carcinoma invasion through the promotion of the epithelial to mesenchymal transition that leads to the acquisition of an invasive phenotype in HEC-1A and RL95-2 cells. Impairment of this initial step with SB-431542, a specific TGF-ß1 inhibitor, precluded further persistent endometrial carcinoma invasion. In conclusion, we showed that the characterization of the molecular changes associated with the acquisition of an aggressive phenotype represents a realistic strategy for the rational identification and characterization of new potential therapeutic targets in an effort to improve the clinical management and the outcome of high-risk endometrial cancer patients.


Subject(s)
Endometrial Neoplasms/genetics , Transforming Growth Factor beta1/physiology , Benzamides/pharmacology , Cell Line, Tumor , Dioxoles/pharmacology , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Epidermal Growth Factor/metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Models, Biological , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Recurrence , Signal Transduction/genetics , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...