Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Publication year range
1.
Cell ; 176(1-2): 98-112.e14, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30633912

ABSTRACT

The ability of circulating tumor cells (CTCs) to form clusters has been linked to increased metastatic potential. Yet biological features and vulnerabilities of CTC clusters remain largely unknown. Here, we profile the DNA methylation landscape of single CTCs and CTC clusters from breast cancer patients and mouse models on a genome-wide scale. We find that binding sites for stemness- and proliferation-associated transcription factors are specifically hypomethylated in CTC clusters, including binding sites for OCT4, NANOG, SOX2, and SIN3A, paralleling embryonic stem cell biology. Among 2,486 FDA-approved compounds, we identify Na+/K+ ATPase inhibitors that enable the dissociation of CTC clusters into single cells, leading to DNA methylation remodeling at critical sites and metastasis suppression. Thus, our results link CTC clustering to specific changes in DNA methylation that promote stemness and metastasis and point to cluster-targeting compounds to suppress the spread of cancer.


Subject(s)
Breast Neoplasms/genetics , Neoplasm Metastasis/genetics , Neoplastic Cells, Circulating/pathology , Animals , Breast Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , DNA Methylation/physiology , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred NOD , Nanog Homeobox Protein/metabolism , Neoplasm Metastasis/physiopathology , Neoplastic Cells, Circulating/metabolism , Octamer Transcription Factor-3/metabolism , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Sin3 Histone Deacetylase and Corepressor Complex
2.
Cell ; 158(5): 1110-1122, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25171411

ABSTRACT

Circulating tumor cell clusters (CTC clusters) are present in the blood of patients with cancer but their contribution to metastasis is not well defined. Using mouse models with tagged mammary tumors, we demonstrate that CTC clusters arise from oligoclonal tumor cell groupings and not from intravascular aggregation events. Although rare in the circulation compared with single CTCs, CTC clusters have 23- to 50-fold increased metastatic potential. In patients with breast cancer, single-cell resolution RNA sequencing of CTC clusters and single CTCs, matched within individual blood samples, identifies the cell junction component plakoglobin as highly differentially expressed. In mouse models, knockdown of plakoglobin abrogates CTC cluster formation and suppresses lung metastases. In breast cancer patients, both abundance of CTC clusters and high tumor plakoglobin levels denote adverse outcomes. Thus, CTC clusters are derived from multicellular groupings of primary tumor cells held together through plakoglobin-dependent intercellular adhesion, and though rare, they greatly contribute to the metastatic spread of cancer.


Subject(s)
Breast Neoplasms/pathology , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/pathology , Animals , Breast Neoplasms/physiopathology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , Sequence Analysis, RNA , Single-Cell Analysis , gamma Catenin/metabolism
3.
Nature ; 607(7917): 156-162, 2022 07.
Article in English | MEDLINE | ID: mdl-35732738

ABSTRACT

The metastatic spread of cancer is achieved by the haematogenous dissemination of circulating tumour cells (CTCs). Generally, however, the temporal dynamics that dictate the generation of metastasis-competent CTCs are largely uncharacterized, and it is often assumed that CTCs are constantly shed from growing tumours or are shed as a consequence of mechanical insults1. Here we observe a striking and unexpected pattern of CTC generation dynamics in both patients with breast cancer and mouse models, highlighting that most spontaneous CTC intravasation events occur during sleep. Further, we demonstrate that rest-phase CTCs are highly prone to metastasize, whereas CTCs generated during the active phase are devoid of metastatic ability. Mechanistically, single-cell RNA sequencing analysis of CTCs reveals a marked upregulation of mitotic genes exclusively during the rest phase in both patients and mouse models, enabling metastasis proficiency. Systemically, we find that key circadian rhythm hormones such as melatonin, testosterone and glucocorticoids dictate CTC generation dynamics, and as a consequence, that insulin directly promotes tumour cell proliferation in vivo, yet in a time-dependent manner. Thus, the spontaneous generation of CTCs with a high proclivity to metastasize does not occur continuously, but it is concentrated within the rest phase of the affected individual, providing a new rationale for time-controlled interrogation and treatment of metastasis-prone cancers.


Subject(s)
Breast Neoplasms , Neoplasm Metastasis , Sleep , Animals , Breast Neoplasms/pathology , Cell Count , Cell Proliferation , Disease Models, Animal , Female , Glucocorticoids , Humans , Insulin , Melatonin , Mice , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/pathology , RNA-Seq , Single-Cell Analysis , Testosterone , Time Factors
4.
Cell ; 144(5): 703-18, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21376233

ABSTRACT

Among breast cancers, triple-negative breast cancer (TNBC) is the most poorly understood and is refractory to current targeted therapies. Using a genetic screen, we identify the PTPN12 tyrosine phosphatase as a tumor suppressor in TNBC. PTPN12 potently suppresses mammary epithelial cell proliferation and transformation. PTPN12 is frequently compromised in human TNBCs, and we identify an upstream tumor-suppressor network that posttranscriptionally controls PTPN12. PTPN12 suppresses transformation by interacting with and inhibiting multiple oncogenic tyrosine kinases, including HER2 and EGFR. The tumorigenic and metastatic potential of PTPN12-deficient TNBC cells is severely impaired upon restoration of PTPN12 function or combined inhibition of PTPN12-regulated tyrosine kinases, suggesting that TNBCs are dependent on the proto-oncogenic tyrosine kinases constrained by PTPN12. Collectively, these data identify PTPN12 as a commonly inactivated tumor suppressor and provide a rationale for combinatorially targeting proto-oncogenic tyrosine kinases in TNBC and other cancers based on their profile of tyrosine-phosphatase activity.


Subject(s)
Breast Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Tumor Suppressor Proteins/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Transformation, Neoplastic , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , MicroRNAs/metabolism , Mutation , Neoplasm Metastasis , Protein Processing, Post-Translational
5.
Nature ; 573(7774): 439-444, 2019 09.
Article in English | MEDLINE | ID: mdl-31485072

ABSTRACT

Metastasis is the major driver of death in patients with cancer. Invasion of surrounding tissues and metastasis have been proposed to initiate following loss of the intercellular adhesion protein, E-cadherin1,2, on the basis of inverse correlations between in vitro migration and E-cadherin levels3. However, this hypothesis is inconsistent with the observation that most breast cancers are invasive ductal carcinomas and express E-cadherin in primary tumours and metastases4. To resolve this discrepancy, we tested the genetic requirement for E-cadherin in metastasis using mouse and human models of both luminal and basal invasive ductal carcinomas. Here we show that E-cadherin promotes metastasis in diverse models of invasive ductal carcinomas. While loss of E-cadherin increased invasion, it also reduced cancer cell proliferation and survival, circulating tumour cell number, seeding of cancer cells in distant organs and metastasis outgrowth. Transcriptionally, loss of E-cadherin was associated with upregulation of genes involved in transforming growth factor-ß (TGFß), reactive oxygen species and apoptosis signalling pathways. At the cellular level, disseminating E-cadherin-negative cells exhibited nuclear enrichment of SMAD2/3, oxidative stress and increased apoptosis. Colony formation of E-cadherin-negative cells was rescued by inhibition of TGFß-receptor signalling, reactive oxygen accumulation or apoptosis. Our results reveal that E-cadherin acts as a survival factor in invasive ductal carcinomas during the detachment, systemic dissemination and seeding phases of metastasis by limiting reactive oxygen-mediated apoptosis. Identifying molecular strategies to inhibit E-cadherin-mediated survival in metastatic breast cancer cells may have potential as a therapeutic approach for breast cancer.


Subject(s)
Antigens, CD , Breast Neoplasms/pathology , Cadherins , Carcinoma, Ductal, Breast/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Animals , Antigens, CD/metabolism , Breast Neoplasms/metabolism , Cadherins/metabolism , Carcinoma, Ductal, Breast/metabolism , Female , Humans , Mice , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism
6.
Nature ; 566(7745): 553-557, 2019 02.
Article in English | MEDLINE | ID: mdl-30728496

ABSTRACT

A better understanding of the features that define the interaction between cancer cells and immune cells is important for the development of new cancer therapies1. However, focus is often given to interactions that occur within the primary tumour and its microenvironment, whereas the role of immune cells during cancer dissemination in patients remains largely uncharacterized2,3. Circulating tumour cells (CTCs) are precursors of metastasis in several types of cancer4-6, and are occasionally found within the bloodstream in association with non-malignant cells such as white blood cells (WBCs)7,8. The identity and function of these CTC-associated WBCs, as well as the molecular features that define the interaction between WBCs and CTCs, are unknown. Here we isolate and characterize individual CTC-associated WBCs, as well as corresponding cancer cells within each CTC-WBC cluster, from patients with breast cancer and from mouse models. We use single-cell RNA sequencing to show that in the majority of these cases, CTCs were associated with neutrophils. When comparing the transcriptome profiles of CTCs associated with neutrophils against those of CTCs alone, we detect a number of differentially expressed genes that outline cell cycle progression, leading to more efficient metastasis formation. Further, we identify cell-cell junction and cytokine-receptor pairs that define CTC-neutrophil clusters, representing key vulnerabilities of the metastatic process. Thus, the association between neutrophils and CTCs drives cell cycle progression within the bloodstream and expands the metastatic potential of CTCs, providing a rationale for targeting this interaction in treatment of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cell Cycle , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/pathology , Neutrophils/pathology , Animals , Breast Neoplasms/therapy , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Exons/genetics , Female , Gene Expression Profiling , Humans , Intercellular Junctions , Mice , Mutation/genetics , Neoplasm Metastasis/genetics , Neoplastic Cells, Circulating/metabolism , Neutrophils/metabolism , Sequence Analysis, RNA , Exome Sequencing
7.
Blood ; 140(2): 99-111, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35468185

ABSTRACT

Cells can use signaling pathway activity over time (ie, dynamics) to control cell fates. However, little is known about the potential existence and function of signaling dynamics in primary hematopoietic stem and progenitor cells (HSPCs). Here, we use time-lapse imaging and tracking of single murine HSPCs from green fluorescent protein-p65/H2BmCherry reporter mice to quantify their nuclear factor κB (NfκB) activity dynamics in response to tumor necrosis factor α and interleukin 1ß. We find response dynamics to be heterogeneous between individual cells, with cell type-specific dynamics distributions. Transcriptome sequencing of single cells physically isolated after live dynamics quantification shows activation of different target gene programs in cells with different dynamics. Finally, artificial induction of oscillatory NfκB activity causes changes in granulocyte/monocyte progenitor behavior. Thus, HSPC behavior can be influenced by signaling dynamics, which are tightly regulated during hematopoietic differentiation and enable cell type-specific responses to the same signaling inputs.


Subject(s)
Hematopoietic Stem Cells , NF-kappa B , Animals , Blood Cells/metabolism , Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , Mice , NF-kappa B/metabolism , Signal Transduction
8.
Br J Cancer ; 128(11): 1981-1990, 2023 06.
Article in English | MEDLINE | ID: mdl-36932192

ABSTRACT

Gastrointestinal (GI) cancers account for 35% of cancer-related deaths, predominantly due to their ability to spread and generate drug-tolerant metastases. Arising from different locations in the GI system, the majority of metastatic GI malignancies colonise the liver and the lungs. In this context, circulating tumour cells (CTCs) are playing a critical role in the formation of new metastases, and their presence in the blood of patients has been correlated with a poor outcome. In addition to their prognostic utility, prospective targeting of CTCs may represent a novel, yet ambitious strategy in the fight against metastasis. A better understanding of CTC biology, mechanistic underpinnings and weaknesses may facilitate the development of previously underappreciated anti-metastasis approaches. Here, along with related clinical studies, we outline a selection of the literature describing biological features of CTCs with an impact on their metastasis forming ability in different GI cancers.


Subject(s)
Gastrointestinal Neoplasms , Neoplastic Cells, Circulating , Humans , Neoplastic Cells, Circulating/pathology , Prospective Studies , Gastrointestinal Neoplasms/pathology , Lung , Prognosis , Neoplasm Metastasis/pathology , Biomarkers, Tumor
9.
J Mammary Gland Biol Neoplasia ; 27(2): 145-153, 2022 06.
Article in English | MEDLINE | ID: mdl-35739379

ABSTRACT

Treatment of metastasis remains a clinical challenge and the majority of breast cancer-related deaths are the result of drug-resistant metastases. The protein tyrosine phosphatase SHP2 encoded by the proto-oncogene PTPN11 promotes breast cancer progression. Inhibition of SHP2 has been shown to decrease metastases formation in various breast cancer models, but specific downstream effectors of SHP2 remain poorly characterized. Certain cytokines in the metastatic cascade facilitate local invasion and promote metastatic colonization. In this study, we investigated cytokines affected by SHP2 that could be relevant for its pro-tumorigenic properties. We used a cytokine array to investigate differentially released cytokines in the supernatant of SHP2 inhibitor-treated breast cancer cells. Expression of CXCL8 transcripts and protein abundance were assessed in human breast cancer cell lines in which we blocked SHP2 using shRNA constructs or an allosteric inhibitor. The impact of SHP2 inhibition on the phospho-tyrosine-proteome and signaling was determined using mass spectrometry. From previously published RNAseq data (Aceto et al. in Nat. Med. 18:529-37, 2012), we computed transcription factor activities using an integrated system for motif activity response analysis (ISMARA) (Balwierz et al. in Genome Res. 24:869-84, 2014). Finally, using siRNA against ETS1, we investigated whether ETS1 directly influences CXCL8 expression levels. We found that IL-8 is one of the most downregulated cytokines in cell supernatants upon SHP2 blockade, with a twofold decrease in CXCL8 transcripts and a fourfold decrease in IL-8 protein. These effects were also observed in preclinical tumor models. Analysis of the phospho-tyrosine-proteome revealed that several effectors of the mitogen-activated protein kinase (MAPK) pathway are downregulated upon SHP2 inhibition in vitro. MEK1/2 inhibition consistently reduced IL-8 levels in breast cancer cell supernatants. Computational analysis of RNAseq data from SHP2-depleted tumors revealed reduced activity of the transcription factor ETS1, a direct target of ERK and a transcription factor reported to regulate IL-8 expression. Our work reveals that SHP2 mediates breast cancer progression by enhancing the production and secretion of the pro-metastatic cytokine IL-8. We also provide mechanistic insights into the effects of SHP2 inhibition and its downstream repercussions. Overall, these results support a rationale for targeting SHP2 in breast cancer.


Subject(s)
Breast Neoplasms , Interleukin-8 , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Proteome , Transcription Factors , Tyrosine
10.
Br J Cancer ; 125(1): 23-27, 2021 07.
Article in English | MEDLINE | ID: mdl-33762721

ABSTRACT

Circulating tumour cell (CTC) clusters have been proposed to be major players in the metastatic spread of breast cancer, particularly during advanced disease stages. Yet, it is unclear whether or not they manifest in early breast cancer, as their occurrence in patients with metastasis-free primary disease has not been thoroughly evaluated. In this study, exploiting nanostructured titanium oxide-coated slides for shear-free CTC identification, we detect clustered CTCs in the curative setting of multiple patients with early breast cancer prior to surgical treatment, highlighting their presence already at early disease stages. These results spotlight an important aspect of metastasis biology and the possibility to intervene with anti-cluster therapeutics already during the early manifestation of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Titanium/chemistry , Breast Neoplasms/surgery , Case-Control Studies , Cell Line, Tumor , Female , Humans , Nanostructures , Neoplasm Metastasis , Neoplasm Staging
11.
Breast Cancer Res Treat ; 188(1): 43-52, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34101078

ABSTRACT

PURPOSE: Therapeutic efficacy of hormonal therapies to target estrogen receptor (ER)-positive breast cancer is limited by the acquisition of ligand-independent ESR1 mutations, which confer treatment resistance to aromatase inhibitors (AIs). Monitoring for the emergence of such mutations may enable individualized therapy. We thus assessed CTC- and ctDNA-based detection of ESR1 mutations with the aim of evaluating non-invasive approaches for the determination of endocrine resistance. PATIENTS AND METHODS: In a prospective cohort of 55 women with hormone receptor-positive metastatic breast cancer, we isolated circulating tumor cells (CTCs) and developed a high-sensitivity method for the detection of ESR1 mutations in these CTCs. In patients with sufficient plasma for the simultaneous extraction of circulating tumor DNA (ctDNA), we performed a parallel analysis of ESR1 mutations using multiplex droplet digital PCR (ddPCR) and examined the agreement between these two platforms. Finally, we isolated single CTCs from a subset of these patients and reviewed RNA expression to explore alternate methods of evaluating endocrine responsiveness. RESULTS: High-sensitivity ESR1 sequencing from CTCs revealed mono- and oligoclonal mutations in 22% of patients. These were concordant with plasma DNA sequencing in 95% of cases. Emergence of ESR1 mutations was correlated both with time to metastatic relapse and duration of AI therapy following such recurrence. The Presence of an ESR1 mutation, compared to ESR1 wild type, was associated with markedly shorter Progression-Free Survival on AI-based therapies (p = 0.0006), but unaltered to other non-AI-based therapies (p = 0.73). Compared with ESR1 mutant cases, AI-resistant CTCs with wild-type ESR1 showed an elevated ER-coactivator RNA signature, consistent with their predicted response to second-line hormonal therapies. CONCLUSION: Blood-based serial monitoring may guide the selection of precision therapeutics for women with AI-resistant ER-positive breast cancer.


Subject(s)
Breast Neoplasms , Circulating Tumor DNA , Neoplastic Cells, Circulating , Estrogen Receptor alpha/genetics , Female , Genotype , Humans , Mutation , Neoplasm Recurrence, Local , Prospective Studies
12.
Recent Results Cancer Res ; 215: 347-368, 2020.
Article in English | MEDLINE | ID: mdl-31605238

ABSTRACT

Next-generation sequencing of DNA and RNA obtained from liquid biopsies of cancer patients may reveal important insights into disease progression and metastasis formation, and it holds the promise to enable new methods for noninvasive screening and clinical decision support. However, implementing liquid biopsy sequencing protocols is challenged by capturing circulating tumor cells or cell-free tumor DNA from blood samples, by amplifying genomic DNA and RNA in a reliable and unbiased manner, and by extracting biologically meaningful signals from the noisy sequencing data. In this chapter, we discuss computational methods for the analysis of DNA and RNA sequencing data obtained from liquid biopsies, addressing these challenges.


Subject(s)
Circulating Tumor DNA/analysis , Circulating Tumor DNA/genetics , High-Throughput Nucleotide Sequencing/methods , Liquid Biopsy , Neoplasms/diagnosis , Neoplasms/genetics , Sequence Analysis, DNA/methods , Sequence Analysis, RNA/methods , Circulating Tumor DNA/blood , Humans
13.
BMC Cancer ; 19(1): 312, 2019 Apr 04.
Article in English | MEDLINE | ID: mdl-30947697

ABSTRACT

BACKGROUND: The tumour microenvironment is a critical regulator of malignant cancer progression. While endothelial cells have been widely studied in the context of tumour angiogenesis, their role as modulators of cancer cell invasion and migration is poorly understood. METHODS: We have investigated the influence of endothelial cells on the invasive and migratory behaviour of human cancer cells in vitro. RESULTS: Upon exposure to culture supernatants of endothelial cells, distinct cancer cells, such as SK-BR-3 cells, showed significantly increased invasion and cell migration concomitant with changes in cell morphology and gene expression reminiscent of an epithelial-mesenchymal transition (EMT). Interestingly, the pro-migratory effect on SK-BR-3 cells was significantly enhanced by supernatants obtained from subconfluent, proliferative endothelial cells rather than from confluent, quiescent endothelial cells. Systematically comparing the supernatants of subconfluent and confluent endothelial cells by quantitative MS proteomics revealed eight candidate proteins that were secreted at significantly higher levels by confluent endothelial cells representing potential inhibitors of cancer cell migration. Among these proteins, nidogen-1 was exclusively expressed in confluent endothelial cells and was found to be necessary and sufficient for the inhibition of SK-BR-3 cell migration. Indeed, SK-BR-3 cells exposed to nidogen-1-depleted endothelial supernatants showed increased promigratory STAT3 phosphorylation along with increased cell migration. This reflects the situation of enhanced SK-BR-3 migration upon stimulation with conditioned medium from subconfluent endothelial cells with inherent absence of nidogen-1 expression. CONCLUSION: The identification of nidogen-1 as an endothelial-derived inhibitor of migration of distinct cancer cell types reveals a novel mechanism of endothelial control over cancer progression.


Subject(s)
Breast Neoplasms/pathology , Endothelial Cells/metabolism , Membrane Glycoproteins/metabolism , Tumor Microenvironment , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Epithelial-Mesenchymal Transition/physiology , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Membrane Glycoproteins/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Primary Cell Culture , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism
14.
Breast Cancer Res ; 20(1): 141, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30458879

ABSTRACT

BACKGROUND: The presence of circulating tumor cells (CTCs) in patients with breast cancer correlates to a bad prognosis. Yet, CTCs are detectable in only a minority of patients with progressive breast cancer, and factors that influence the abundance of CTCs remain elusive. METHODS: We conducted CTC isolation and enumeration in a selected group of 73 consecutive patients characterized by progressive invasive breast cancer, high tumor load and treatment discontinuation at the time of CTC isolation. CTCs were quantified with the Parsortix microfluidic device. Clinicopathological variables, blood counts at the time of CTC isolation and detailed treatment history prior to blood sampling were evaluated for each patient. RESULTS: Among 73 patients, we detected at least one CTC per 7.5 ml of blood in 34 (46%). Of these, 22 (65%) had single CTCs only, whereas 12 (35%) featured both single CTCs and CTC clusters. Treatment with the monoclonal antibody denosumab correlated with the absence of CTCs, both when considering all patients and when considering only those with bone metastasis. We also found that low red blood cell count was associated with the presence of CTCs, whereas high CA 15-3 tumor marker, high mean corpuscular volume, high white blood cell count and high mean platelet volume associated specifically with CTC clusters. CONCLUSIONS: In addition to blood count correlatives to single and clustered CTCs, we found that denosumab treatment associates with most patients lacking CTCs from their peripheral circulation. Prospective studies will be needed to validate the involvement of denosumab in the prevention of CTC generation.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Denosumab/pharmacology , Erythrocytes , Neoplastic Cells, Circulating/drug effects , Aged , Antineoplastic Agents/therapeutic use , Breast/pathology , Breast Neoplasms/blood , Breast Neoplasms/pathology , Cell Count/methods , Denosumab/therapeutic use , Disease Progression , Female , Humans , Microfluidic Analytical Techniques/methods , Middle Aged , Neoplasm Invasiveness/pathology , Prognosis , Retrospective Studies
15.
Br J Cancer ; 119(4): 487-491, 2018 08.
Article in English | MEDLINE | ID: mdl-30065256

ABSTRACT

Human glioblastoma (GBM) is a highly aggressive, invasive and hypervascularised malignant brain cancer. Individual circulating tumour cells (CTCs) are sporadically found in GBM patients, yet it is unclear whether multicellular CTC clusters are generated in this disease and whether they can bypass the physical hurdle of the blood-brain barrier.  Here, we assessed CTC presence and composition at multiple time points in 13 patients with progressing GBM during an open-label phase 1/2a study with the microtubule inhibitor BAL101553. We observe CTC clusters ranging from 2 to 23 cells and present at multiple sampling time points in a GBM patient with pleomorphism and extensive necrosis, throughout disease progression. Exome sequencing of GBM CTC clusters highlights variants in 58 cancer-associated genes including ATM, PMS2, POLE, APC, XPO1, TFRC, JAK2, ERBB4 and ALK. Together, our findings represent the first evidence of the presence of CTC clusters in GBM.


Subject(s)
Benzimidazoles/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplastic Cells, Circulating/pathology , Oxadiazoles/administration & dosage , Animals , Benzimidazoles/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Count , Cluster Analysis , Disease Progression , Female , Gene Regulatory Networks/drug effects , Genetic Variation , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Mice , Mutation , Neoplastic Cells, Circulating/chemistry , Neoplastic Cells, Circulating/drug effects , Oxadiazoles/pharmacology , Exome Sequencing , Xenograft Model Antitumor Assays
16.
Nat Methods ; 12(7): 685-91, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25984697

ABSTRACT

Cancer cells metastasize through the bloodstream either as single migratory circulating tumor cells (CTCs) or as multicellular groupings (CTC clusters). Existing technologies for CTC enrichment are designed to isolate single CTCs, and although CTC clusters are detectable in some cases, their true prevalence and significance remain to be determined. Here we developed a microchip technology (the Cluster-Chip) to capture CTC clusters independently of tumor-specific markers from unprocessed blood. CTC clusters are isolated through specialized bifurcating traps under low-shear stress conditions that preserve their integrity, and even two-cell clusters are captured efficiently. Using the Cluster-Chip, we identified CTC clusters in 30-40% of patients with metastatic breast or prostate cancer or with melanoma. RNA sequencing of CTC clusters confirmed their tumor origin and identified tissue-derived macrophages within the clusters. Efficient capture of CTC clusters will enable the detailed characterization of their biological properties and role in metastasis.


Subject(s)
Microfluidic Analytical Techniques , Neoplastic Cells, Circulating , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Male , Prostatic Neoplasms/pathology , Sequence Analysis, RNA
17.
J Neurooncol ; 135(3): 487-496, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28852935

ABSTRACT

SHP2 is a cytoplasmic protein tyrosine phosphatase (PTPase) involved in multiple signaling pathways and was the first identified proto-oncogene PTPase. Previous work in glioblastoma (GBM) has demonstrated the role of SHP2 PTPase activity in modulating the oncogenic phenotype of adherent GBM cell lines. Mutations in PTPN11, the gene encoding SHP2, have been identified with increasing frequency in GBM. Given the importance of SHP2 in developing neural stem cells, and the importance of glioma stem cells (GSCs) in GBM oncogenesis, we explored the functional role of SHP2 in GSCs. Using paired differentiated and stem cell primary cultures, we investigated the association of SHP2 expression with the tumor stem cell compartment. Proliferation and soft agar assays were used to demonstrate the functional contribution of SHP2 to cell growth and transformation. SHP2 expression correlated with SOX2 expression in GSC lines and was decreased in differentiated cells. Forced differentiation of GSCs by removal of growth factors, as confirmed by loss of SOX2 expression, also resulted in decreased SHP2 expression. Lentiviral-mediated knockdown of SHP2 inhibited proliferation. Finally, growth in soft-agar was similarly inhibited by loss of SHP2 expression. Our results show that SHP2 function is required for cell growth and transformation of the GSC compartment in GBM.


Subject(s)
Brain Neoplasms/enzymology , Carcinogenesis/metabolism , Cell Proliferation/physiology , Glioma/enzymology , Neoplastic Stem Cells/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Adult , Aged , Brain Neoplasms/pathology , Carcinogenesis/pathology , Cell Culture Techniques , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioma/pathology , Humans , Male , Neoplastic Stem Cells/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Mas , SOXB1 Transcription Factors/metabolism , Tissue Scaffolds
19.
Mol Oncol ; 18(1): 3-5, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37899655

ABSTRACT

The metastatic process is an extraordinarily complex step-by-step procedure, characterized by many analogies with migratory patterns of humans or animals across our planet. The ongoing interrogation of circulating tumor cells (CTCs), caught in the act of spreading from one location to another, is revealing distinct behaviors including biological, physical, and mechanical features that impact on their likelihood to form metastasis. In this viewpoint, I will discuss some of these findings and provide a perspective on the metastatic journey, open questions and opportunities to exploit some of the most recent discoveries for the development of antimetastasis medicines.


Subject(s)
Neoplastic Cells, Circulating , Animals , Humans , Neoplastic Cells, Circulating/pathology , Neoplasm Metastasis
20.
Cancer Discov ; 14(6): 920-933, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38581442

ABSTRACT

Circulating tumor cells (CTCs) play a pivotal role in metastasis, the leading cause of cancer-associated death. Recent improvements of CTC isolation tools, coupled with a steady development of multiomics technologies at single-cell resolution, have enabled an extensive exploration of CTC biology, unlocking insights into their molecular profiles. A detailed molecular portrait requires CTC interrogation across various levels encompassing genomic, epigenetic, transcriptomic, proteomic and metabolic features. Here, we review how state-of-the-art multiomics applied to CTCs are shedding light on how cancer spreads. Further, we highlight the potential implications of CTC profiling for clinical applications aimed at enhancing cancer diagnosis and treatment. SIGNIFICANCE: Exploring the complexity of cancer progression through cutting-edge multiomics studies holds the promise of uncovering novel aspects of cancer biology and identifying therapeutic vulnerabilities to suppress metastasis.


Subject(s)
Neoplasms , Neoplastic Cells, Circulating , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Humans , Neoplasms/pathology , Neoplasms/genetics , Neoplasms/blood , Neoplasms/metabolism , Genomics/methods , Proteomics/methods , Biomarkers, Tumor , Single-Cell Analysis/methods , Multiomics
SELECTION OF CITATIONS
SEARCH DETAIL