Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.823
Filtrer
1.
Sci Rep ; 14(1): 18189, 2024 08 06.
Article de Anglais | MEDLINE | ID: mdl-39107343

RÉSUMÉ

Desmosomes are intercellular adhesion complexes providing mechanical coupling and tissue integrity. Previously, a correlation of desmosomal molecule expression with invasion and metastasis formation in several tumor entities was described together with a relevance for circulating tumor cell cluster formation. Here, we investigated the contribution of the desmosomal core adhesion molecule desmoglein-2 (DSG2) to the initial steps of liver metastasis formation by pancreatic cancer cells using a novel ex vivo liver perfusion mouse model. We applied the pancreatic ductal adenocarcinoma cell line AsPC-1 with and without a knockout (KO) of DSG2 and generated mouse lines with a hepatocyte-specific KO of the known interacting partners of DSG2 (DSG2 and desmocollin-2). Liver perfusion with DSG2 KO AsPC-1 cells led to smaller circulating cell clusters and a reduced number of cells adhering to murine livers compared to control cells. While this was independent of the expression levels of desmosomal adhesion molecules in hepatocytes, we show that increased cluster size of cancer cells, which correlates with stronger cell-cell adhesion and expression of desmosomal molecules, is a major factor contributing to the early phase of metastatic spreading. In conclusion, impaired desmosomal adhesion results in reduced circulating cell cluster size, which is relevant for seeding and attachment of metastatic cells to the liver.


Sujet(s)
Adhérence cellulaire , Desmogléine-2 , Desmosomes , Tumeurs du foie , Tumeurs du pancréas , Animaux , Desmosomes/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Souris , Tumeurs du foie/secondaire , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Lignée cellulaire tumorale , Humains , Desmogléine-2/métabolisme , Desmogléine-2/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/génétique , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Souris knockout , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie
2.
BMC Cancer ; 24(1): 1016, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39148033

RÉSUMÉ

BACKGROUND: Triple negative breast cancer (TNBC) is an aggressive subtype with poor prognosis. We aimed to determine whether circulating tumor DNA (ctDNA) and circulating tumor cell (CTC) could predict response and long-term outcomes to neoadjuvant chemotherapy (NAC). METHODS: Patients with TNBC were enrolled between 2017-2021 at The University of Texas MD Anderson Cancer Center (Houston, TX). Serial plasma samples were collected at four timepoints: pre-NAC (baseline), 12-weeks after NAC (mid-NAC), after NAC/prior to surgery (post-NAC), and one-year after surgery. ctDNA was quantified using a tumor-informed ctDNA assay (SignateraTM, Natera, Inc.) and CTC enumeration using CellSearch. Wilcoxon and Fisher's exact tests were used for comparisons between groups and Kaplan-Meier analysis used for survival outcomes. RESULTS: In total, 37 patients were enrolled. The mean age was 50 and majority of patients had invasive ductal carcinoma (34, 91.9%) with clinical T2, (25, 67.6%) node-negative disease (21, 56.8%). Baseline ctDNA was detected in 90% (27/30) of patients, of whom 70.4% (19/27) achieved ctDNA clearance by mid-NAC. ctDNA clearance at mid-NAC was significantly associated with pathologic complete response (p = 0.02), whereas CTC clearance was not (p = 0.52). There were no differences in overall survival (OS) and recurrence-free survival (RFS) with positive baseline ctDNA and CTC. However, positive ctDNA at mid-NAC was significantly associated with worse OS and RFS (p = 0.0002 and p = 0.0034, respectively). CONCLUSIONS: Early clearance of ctDNA served as a predictive and prognostic marker in TNBC. Personalized ctDNA monitoring during NAC may help predict response and guide treatment.


Sujet(s)
ADN tumoral circulant , Traitement néoadjuvant , Cellules tumorales circulantes , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/sang , Tumeurs du sein triple-négatives/mortalité , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , ADN tumoral circulant/sang , ADN tumoral circulant/génétique , Femelle , Traitement néoadjuvant/méthodes , Adulte d'âge moyen , Adulte , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Marqueurs biologiques tumoraux/sang , Sujet âgé , Pronostic , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Résultat thérapeutique
4.
Drug Resist Updat ; 76: 101119, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39111134

RÉSUMÉ

Cancer metastasis and therapy resistance are intricately linked with the dynamics of Epithelial-Mesenchymal Transition (EMT) and Circulating Tumor Cells (CTCs). EMT hybrid cells, characterized by a blend of epithelial and mesenchymal traits, have emerged as pivotal in metastasis and demonstrate remarkable plasticity, enabling transitions across cellular states crucial for intravasation, survival in circulation, and extravasation at distal sites. Concurrently, CTCs, which are detached from primary tumors and travel through the bloodstream, are crucial as potential biomarkers for cancer prognosis and therapeutic response. There is a significant interplay between EMT hybrid cells and CTCs, revealing a complex, bidirectional relationship that significantly influences metastatic progression and has a critical role in cancer drug resistance. This resistance is further influenced by the tumor microenvironment, with factors such as tumor-associated macrophages, cancer-associated fibroblasts, and hypoxic conditions driving EMT and contributing to therapeutic resistance. It is important to understand the molecular mechanisms of EMT, characteristics of EMT hybrid cells and CTCs, and their roles in both metastasis and drug resistance. This comprehensive understanding sheds light on the complexities of cancer metastasis and opens avenues for novel diagnostic approaches and targeted therapies and has significant advancements in combating cancer metastasis and overcoming drug resistance.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Transition épithélio-mésenchymateuse , Métastase tumorale , Tumeurs , Cellules tumorales circulantes , Microenvironnement tumoral , Humains , Cellules tumorales circulantes/effets des médicaments et des substances chimiques , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Animaux , Marqueurs biologiques tumoraux/métabolisme , Cellules M
5.
Adv Surg ; 58(1): 135-142, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39089773

RÉSUMÉ

The notion that technically resectable pancreatic ductal adenocarcinoma presents as localized disease is now known to be inaccurate. Evidence supports that most patients have subclinical systemic dissemination at the time of diagnosis. It is now widely accepted that both a local and systemic component of disease coexist, each requiring treatment of improved survival and potential cure. The advent of multiagent chemotherapy regimens has resulted in a modest improvement in survival. Consequently, this article will emphasize the expanding potential and significance of circulating tumor cells in the prognostication and management of patients with pancreatic cancer.


Sujet(s)
Carcinome du canal pancréatique , Cellules tumorales circulantes , Tumeurs du pancréas , Humains , Cellules tumorales circulantes/anatomopathologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/sang , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/mortalité , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/sang , Carcinome du canal pancréatique/mortalité , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/chirurgie , Pronostic
7.
Sci Rep ; 14(1): 15019, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38951573

RÉSUMÉ

Circulating tumor cells (CTCs) represent a rare and heterogeneous population of cancer cells that are detached from the tumor site and entered blood or lymphatic circulation. Once disseminated in distant tissues, CTCs could remain dormant or create a tumor mass causing serious danger for patients. Many technologies exist to isolate CTCs from patients' blood samples, mostly based on microfluidic systems or by sorting them according to their surface antigens, notably EpCAM, and/or cytokeratins for carcinoma. ScreenCell has developed an easy-to-use, antigen-independent, rapid, cost-effective, and efficient technology that isolates CTCs according to their bigger size compared to the blood cells. This study provides the technical information necessary to isolate and characterize CTCs from mouse blood. By using blood samples from transgenic mice with breast cancer or from WT mice in which we spiked cancer cells, we showed that ScreenCell technology is compatible with standard EDTA blood collection tubes. Furthermore, the ScreenCell Cyto kit could treat up to 500 µl and the ScreenCell MB kit up to 200 µl of mouse blood. As the ScreenCell MB kit captures unaltered live CTCs, we have shown that their DNA could be efficiently extracted, and the isolated cells could be grown in culture. In conclusion, ScreenCell provides a rapid, easy, antigen-independent, cost-effective, and efficient technology to isolate and characterize CTCs from the blood samples of cancer patients and murine models. Thanks to this technology CTCs could be captured fixed or alive. Murine cancer models are extensively used in pre-clinical studies. Therefore, this study demonstrates the crucial technical points necessary while manipulating mouse blood samples using ScreenCell technology.


Sujet(s)
Séparation cellulaire , Souris transgéniques , Cellules tumorales circulantes , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Animaux , Souris , Séparation cellulaire/méthodes , Femelle , Humains , Lignée cellulaire tumorale , Tumeurs du sein/anatomopathologie , Tumeurs du sein/sang
8.
Nat Commun ; 15(1): 5849, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38992001

RÉSUMÉ

The effective isolation of rare target cells, such as circulating tumor cells, from whole blood is still challenging due to the lack of a capturing surface with strong target-binding affinity and non-target-cell resistance. Here we present a solution leveraging the flexibility of bacterial virus (phage) nanofibers with their sidewalls displaying target circulating tumor cell-specific aptamers and their ends tethered to magnetic beads. Such flexible phages, with low stiffness and Young's modulus, can twist and adapt to recognize the cell receptors, energetically enhancing target cell capturing and entropically discouraging non-target cells (white blood cells) adsorption. The magnetic beads with flexible phages can isolate and count target cells with significant increase in cell affinity and reduction in non-target cell absorption compared to magnetic beads having rigid phages. This differentiates breast cancer patients and healthy donors, with impressive area under the curve (0.991) at the optimal detection threshold (>4 target cells mL-1). Immunostaining of captured circulating tumor cells precisely determines breast cancer subtypes with a diagnostic accuracy of 91.07%. Our study reveals the power of viral mechanical attributes in designing surfaces with superior target binding and non-target anti-fouling.


Sujet(s)
Tumeurs du sein , Cellules tumorales circulantes , Humains , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie , Tumeurs du sein/virologie , Femelle , Aptamères nucléotidiques/métabolisme , Nanofibres/composition chimique , Lignée cellulaire tumorale , Bactériophages/génétique
9.
Cells ; 13(14)2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-39056749

RÉSUMÉ

BACKGROUND: Metastasis is the main cause of cancer-related deaths, but efficient targeted therapies against metastasis are still missing. Major gaps exist in our understanding of the metastatic cascade, as existing methods cannot combine sensitivity, robustness, and practicality to dissect cancer progression. Addressing this issue requires improved strategies to distinguish early metastatic colonization from metastatic outgrowth. METHODS: Luciferase-labelled MDA-MB-231, MCF7, and 4T1 breast cancer cells were spiked into samples from tumour-naïve mice to establish the limit of detection for disseminated tumour cells. Luciferase-labelled breast cancer cells (±unlabelled cancer-associated fibroblasts; CAFs) were orthotopically implanted in immunocompromised mice. An ex vivo luciferase assay was used to quantify tumour cell dissemination. RESULTS: In vitro luciferase assay confirmed a linear and positive correlation between cancer cell numbers and the bioluminescence detected at single cell level in blood, brain, lung, liver, and mammary fat pad samples. Remarkably, single luciferase-labelled cancer cells were detectable in all of these sites, as the bioluminescence quantified in the analysed samples was substantially higher than background levels. Ex vivo, circulating tumour cells, metastasis, and tumour self-seeding were detected in all samples from animals implanted with highly metastatic luciferase-labelled MDA-MB-231 cells. In turn, detection of poorly metastatic luciferase-labelled MCF7 cells was scarce but significantly enhanced upon co-implantation with CAFs as early as 20 days after the experiment was initiated. CONCLUSIONS: These results demonstrate the feasibility of using an ultrasensitive luciferase-based method to dissect the mechanisms of early metastatic colonization to improving the development of antimetastatic therapies.


Sujet(s)
Tumeurs du sein , Métastase tumorale , Cellules tumorales circulantes , Animaux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/sang , Femelle , Souris , Humains , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Modèles animaux de maladie humaine , Lignée cellulaire tumorale , Dépistage précoce du cancer/méthodes , Luciferases/métabolisme
10.
Anticancer Res ; 44(8): 3481-3491, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39060051

RÉSUMÉ

BACKGROUND/AIM: Precision medicine aims to revolutionize healthcare by tailoring treatment regimens. This study aimed to integrate comprehensive tumor genomic profiling (CTGP) by targeted-gene panel sequencing and drug screening by circulating tumor cell-derived organoids (CTOs) into clinical practice for the treatment of gastrointestinal (GI) cancers. PATIENTS AND METHODS: Nine patients with various GI cancers underwent CTGP and CTO drug sensitivity testing. CTGP results guided targeted therapy and immunotherapy, while CTO drug sensitivity predicted response to chemotherapy and targeted agents. The drug recommendations from two platforms were correlated with the treatment response to the suggested medications retrospectively. RESULTS: Five patients received therapies aligned with CTGP, including HER2-targeted treatment, immunotherapy, and BRAF/MEK dual inhibition, showing positive responses. CTO drug sensitivity predicted progression under regorafenib (low potential benefit) and good response to chemotherapy with high potential benefit. The combination of CTGP and CTO drug sensitivity may exhibit significant correlation with clinical outcomes during treatment with candidate drugs, demonstrating a sensitivity of 79% and an accuracy of 75%. This encompasses various treatment modalities, such as chemotherapy, targeted therapy, and immunotherapy. CONCLUSION: The present investigation elucidated the integration of CTGP and CTO drug sensitivity screening into clinical practice in a complementary manner, showcasing a significant correlation between treatment response and testing outcomes. Additional prospective evaluation of these two testing modalities in a large cohort is warranted to confirm whether the inclusion of CTO drug sensitivity screening confers enhanced survival benefits compared to utilizing CTGP alone.


Sujet(s)
Tumeurs gastro-intestinales , Cellules tumorales circulantes , Organoïdes , Humains , Tumeurs gastro-intestinales/génétique , Tumeurs gastro-intestinales/traitement médicamenteux , Tumeurs gastro-intestinales/anatomopathologie , Tumeurs gastro-intestinales/sang , Femelle , Mâle , Organoïdes/anatomopathologie , Organoïdes/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Sujet âgé , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/effets des médicaments et des substances chimiques , Médecine de précision/méthodes , Génomique/méthodes , Études rétrospectives , Analyse de profil d'expression de gènes/méthodes , Marqueurs biologiques tumoraux/génétique , Thérapie moléculaire ciblée/méthodes , Adulte , Tests de criblage d'agents antitumoraux/méthodes
11.
Anticancer Res ; 44(8): 3317-3319, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39060063

RÉSUMÉ

Breast cancer, a multifaceted disease, presents a dynamic ecosystem where the primary tumor interacts intricately with its microenvironment, circulatory system, and distant organs. Circulating tumor cells (CTCs) disseminate from the primary tumor to organs, such as the brain, lungs, liver, and bones, encountering various fates: cell death, cellular dormancy, or senescence. Dormant cells, characterized by reversible growth arrest at the G0/G1 phase of the cell cycle, pose a significant challenge as they evade conventional treatments and can later reawaken, leading to cancer relapse. The phenomenon of tumor dormancy is influenced by the tumor microenvironment, immune modulation, and cellular adaptations. Emerging evidence suggests that breast-conserving surgery coupled with radiation therapy offers superior survival benefits compared to mastectomy, potentially due to the 'breast homing phenomenon.' This hypothesis posits that residual breast tissue provides a niche for reactivated dormant cells, reducing distant metastasis. Immunotherapy and lifestyle modifications, including diet and exercise, show promise in managing dormant cells. Understanding the mechanisms of dormancy and developing targeted therapies are crucial for achieving long-term remission and potentially curing breast cancer.


Sujet(s)
Tumeurs du sein , Microenvironnement tumoral , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/thérapie , Tumeurs du sein/métabolisme , Femelle , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme
12.
Biomolecules ; 14(7)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-39062471

RÉSUMÉ

Circulating tumor cells (CTCs) are some of the key culprits that cause cancer metastasis and metastasis-related deaths. These cells exist in a dynamic microenvironment where they experience fluid shear stress (FSS), and the CTCs that survive FSS are considered to be highly metastatic and stem cell-like. Biophysical stresses such as FSS are also known to cause the production of extracellular vesicles (EVs) that can facilitate cell-cell communication by carrying biomolecular cargos such as microRNAs. Here, we hypothesized that physiological FSS will impact the yield of EV production, and that these EVs will have biomolecules that transform the recipient cells. The EVs were isolated using direct flow filtration with and without FSS from the MDA-MB-231 cancer cell line, and the expression of key stemness-related genes and microRNAs was characterized. There was a significantly increased yield of EVs under FSS. These EVs also contained significantly increased levels of miR-21, which was previously implicated to promote metastatic progression and chemotherapeutic resistance. When these EVs from FSS were introduced to MCF-7 cancer cells, the recipient cells had a significant increase in their stem-like gene expression and CD44+/CD24- cancer stem cell-like subpopulation. There was also a correlated increased proliferation along with an increased ATP production. Together, these findings indicate that the presence of physiological FSS can directly influence the EVs' production and their contents, and that the EV-mediated transfer of miR-21 can have an important role in FSS-existing contexts, such as in cancer metastasis.


Sujet(s)
Tumeurs du sein , Vésicules extracellulaires , microARN , Cellules souches tumorales , Humains , Vésicules extracellulaires/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Femelle , microARN/génétique , microARN/métabolisme , Cellules MCF-7 , Lignée cellulaire tumorale , Contrainte mécanique , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie , Régulation de l'expression des gènes tumoraux , Phénotype , Antigènes CD24/métabolisme , Antigènes CD24/génétique
13.
Biomolecules ; 14(7)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39062515

RÉSUMÉ

Gliomas are the most common type of malignant brain tumor and are characterized by a plethora of heterogeneous molecular alterations. Current treatments require the emergence of reliable biomarkers that will aid personalized treatment decisions and increase life expectancy. Glioma tissues are not as easily accessible as other solid tumors; therefore, detecting prominent biomarkers in biological fluids is necessary. Cerebrospinal fluid (CSF) circulates adjacent to the cerebral parenchyma and holds promise for discovering useful prognostic, diagnostic, and predictive biomarkers. In this review, we summarize extensive research regarding the role of circulating DNA, tumor cells, proteins, microRNAs, metabolites, and extracellular vesicles as potential CSF biomarkers for glioma diagnosis, prognosis, and monitoring. Future studies should address discrepancies and issues of specificity regarding CSF biomarkers, as well as the validation of candidate biomarkers.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du cerveau , Vésicules extracellulaires , Gliome , Humains , Gliome/liquide cérébrospinal , Gliome/diagnostic , Marqueurs biologiques tumoraux/liquide cérébrospinal , Tumeurs du cerveau/liquide cérébrospinal , Tumeurs du cerveau/diagnostic , Vésicules extracellulaires/métabolisme , microARN/liquide cérébrospinal , Pronostic , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie
14.
Int J Mol Sci ; 25(14)2024 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-39063215

RÉSUMÉ

Gliomas, particularly glioblastoma (GBM), represent the most prevalent and aggressive tumors of the central nervous system (CNS). Despite recent treatment advancements, patient survival rates remain low. The diagnosis of GBM traditionally relies on neuroimaging methods such as magnetic resonance imaging (MRI) or computed tomography (CT) scans and postoperative confirmation via histopathological and molecular analysis. Imaging techniques struggle to differentiate between tumor progression and treatment-related changes, leading to potential misinterpretation and treatment delays. Similarly, tissue biopsies, while informative, are invasive and not suitable for monitoring ongoing treatments. These challenges have led to the emergence of liquid biopsy, particularly through blood samples, as a promising alternative for GBM diagnosis and monitoring. Presently, blood and cerebrospinal fluid (CSF) sampling offers a minimally invasive means of obtaining tumor-related information to guide therapy. The idea that blood or any biofluid tests can be used to screen many cancer types has huge potential. Tumors release various components into the bloodstream or other biofluids, including cell-free nucleic acids such as microRNAs (miRNAs), circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), proteins, extracellular vesicles (EVs) or exosomes, metabolites, and other factors. These factors have been shown to cross the blood-brain barrier (BBB), presenting an opportunity for the minimally invasive monitoring of GBM as well as for the real-time assessment of distinct genetic, epigenetic, transcriptomic, proteomic, and metabolomic changes associated with brain tumors. Despite their potential, the clinical utility of liquid biopsy-based circulating biomarkers is somewhat constrained by limitations such as the absence of standardized methodologies for blood or CSF collection, analyte extraction, analysis methods, and small cohort sizes. Additionally, tissue biopsies offer more precise insights into tumor morphology and the microenvironment. Therefore, the objective of a liquid biopsy should be to complement and enhance the diagnostic accuracy and monitoring of GBM patients by providing additional information alongside traditional tissue biopsies. Moreover, utilizing a combination of diverse biomarker types may enhance clinical effectiveness compared to solely relying on one biomarker category, potentially improving diagnostic sensitivity and specificity and addressing some of the existing limitations associated with liquid biomarkers for GBM. This review presents an overview of the latest research on circulating biomarkers found in GBM blood or CSF samples, discusses their potential as diagnostic, predictive, and prognostic indicators, and discusses associated challenges and future perspectives.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du cerveau , Glioblastome , Cellules tumorales circulantes , Humains , Glioblastome/diagnostic , Glioblastome/sang , Glioblastome/anatomopathologie , Biopsie liquide/méthodes , Marqueurs biologiques tumoraux/sang , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie , Tumeurs du cerveau/diagnostic , Tumeurs du cerveau/sang , Tumeurs du cerveau/anatomopathologie , ADN tumoral circulant/sang , ADN tumoral circulant/liquide cérébrospinal
15.
ACS Appl Mater Interfaces ; 16(28): 36106-36116, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38955781

RÉSUMÉ

Accurate detection of heterogeneous circulating tumor cells (CTCs) is critical as they can make tumor cells more aggressive, drug-resistant, and metastasizing. Although the leukocyte membrane coating strategy is promising in meeting the challenge of detecting heterogeneous CTCs due to its inherent antiadhesive properties, it is still limited by the reduction or loss of expression of known markers. Bioorthogonal glycol-metabolic engineering is expected to break down this barrier by feeding the cells with sugar derivatives with a unique functional group to establish artificial targets on the surface of tumor cells. Herein, an engineered leukocyte biomimetic colorimetric sensor was accordingly fabricated for high-efficient detection of heterogeneous CTCs. Compared with conventional leukocyte membrane coating, the sensor could covalently bound to the heterogeneous CTCs models fed with Ac4ManNAz in vitro through the synergy of bioorthogonal chemistry and metabolic glycoengineering, ignoring the phenotypic changes of heterogeneous CTCs. Meanwhile, a sandwich structure composed of leukocyte biomimetic layer/CTCs/MoS2 nanosheet was formed for visual detection of HeLa cells as low as 10 cells mL-1. Overall, this approach can overcome the dependence of conventional cell membrane biomimetic technology on specific cell phenotypes and provide a new viewpoint to highly efficiently detect heterogeneous CTCs.


Sujet(s)
Matériaux biomimétiques , Colorimétrie , Leucocytes , Cellules tumorales circulantes , Humains , Colorimétrie/méthodes , Cellules HeLa , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Leucocytes/cytologie , Leucocytes/métabolisme , Matériaux biomimétiques/composition chimique , Biomimétique/méthodes , Techniques de biocapteur/méthodes
16.
J Hematol Oncol ; 17(1): 55, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39075488

RÉSUMÉ

Liquid biopsy, an advanced technology for analyzing body fluid samples, is gaining traction in cancer diagnostics and monitoring. Blood-based liquid biopsy, particularly focusing on cell-free DNAs (cf-DNAs), circulating tumor cells (CTCs), and extracellular vesicles (EVs), has garnered significant attention. EVs stand out for their potential in tumor diagnosis, prognosis prediction, and treatment response assessment, owing to their stable molecular cargo and clear extraction process. At the recent American Association for Cancer Research (AACR) Annual Meeting 2024, groundbreaking EVs-based liquid biopsy studies showcased promising strides in early detection and diagnosis of various cancers, including breast cancer (BC), high-grade serous ovarian cancer (HGSOC), pancreatic ductal adenocarcinoma (PDAC), colorectal cancer (CRC), colon adenocarcinoma (COAD), head and neck cancer (HNC), neuroblastoma, and retinoblastoma (RB). Despite these advancements, challenges persist in translating EVs biomarkers into clinical practice. Overcoming these challenges promises to propel EVs-based liquid biopsy into a new era of personalized precision medicine, revolutionizing cancer detection, monitoring, and treatment.


Sujet(s)
Marqueurs biologiques tumoraux , Vésicules extracellulaires , Tumeurs , Cellules tumorales circulantes , Humains , Biopsie liquide/méthodes , Vésicules extracellulaires/métabolisme , Tumeurs/diagnostic , Marqueurs biologiques tumoraux/analyse , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Médecine de précision/méthodes
17.
Cell Rep ; 43(7): 114512, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39003738

RÉSUMÉ

Tumor self-seeding is a process whereby circulating tumor cells (CTCs) recolonize the primary tumor, which promotes tumor growth, angiogenesis, and invasion. However, the detailed nature and functions of tumor self-seeded cells (TSCs) have not been well defined due to challenges in tracking and isolating TSCs. Here, we report an accurate animal model using photoconvertible tagging to recapitulate the spontaneous process of tumor self-seeding and identify TSCs as a subpopulation of primary tumor cells with enhanced invasiveness and survival. We demonstrate transmembrane-4-L-six-family-1 (TM4SF1) as a marker of TSCs, which promotes migration, invasion, and anchorage-independent survival in cancer cells. By analyzing single-cell RNA sequencing datasets, we identify a potential TSC population with a metastatic profile in patients with cancer, which is detectable in early-stage disease and expands during cancer progression. In summary, we establish a framework to study TSCs and identify emerging cell targets with diagnostic, prognostic, or therapeutic potential in cancers.


Sujet(s)
Cellules tumorales circulantes , Humains , Animaux , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie , Souris , Lignée cellulaire tumorale , Mouvement cellulaire , Protéines tumorales/métabolisme , Protéines tumorales/génétique , Invasion tumorale , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Tumeurs/génétique , Marqueurs biologiques tumoraux/métabolisme , Antigènes de surface
18.
Sci Rep ; 14(1): 17501, 2024 07 30.
Article de Anglais | MEDLINE | ID: mdl-39080445

RÉSUMÉ

Circulating tumor cells (CTCs) are precursors of cancer in the blood and provide an attractive source for dynamic monitoring of disease progression and tumor heterogeneity. However, the scarcity of CTCs in the bloodstream has limited their use in clinical practice. In this study, we present a workflow for easy detection of CTCs by cytokeratin staining using the FDA-cleared Parsortix device for size-based microfluidic enrichment. To minimize sample handling, the isolated cells are stained inside the separation cassette and harvested for subsequent single cell isolation and whole genome copy-number analysis. We validated the workflow on a panel of four prostate cancer cell lines spiked into healthy donor blood collected in CellRescue or EDTA tubes, resulting in mean recoveries of 42% (16-69%). Furthermore, we evaluated the clinical utility in a cohort of 12 metastatic prostate cancer patients and found CTCs in 67% of patients ranging from 0 to 1172 CTCs in 10 mL blood. Additionally, we isolated single patient-derived CTCs and identified genomic aberrations associated with treatment response and clinical outcome. Thus, this workflow provides a readily scalable strategy for analysis of single CTCs, applicable for use in monitoring studies to identify genomic variations important for guiding clinical therapy decision.


Sujet(s)
Cellules tumorales circulantes , Tumeurs de la prostate , Analyse sur cellule unique , Flux de travaux , Humains , Mâle , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/sang , Analyse sur cellule unique/méthodes , Lignée cellulaire tumorale , Métastase tumorale , Séparation cellulaire/méthodes , Techniques d'analyse microfluidique/instrumentation , Techniques d'analyse microfluidique/méthodes , Microfluidique/méthodes , Coloration et marquage/méthodes , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/sang
19.
World J Urol ; 42(1): 454, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39073634

RÉSUMÉ

BACKGROUND: The morphology of tumor thrombus varies from person to person and it may affect surgical methods and tumor prognosis. However, studies on the morphology of tumor thrombus are limited. The purpose of our study was to evaluate the impact of tumor thrombus morphology on surgical complexity. METHODS: We retrospectively reviewed the clinical data of 229 patients with renal cell carcinoma combined with inferior vena cava (IVC) tumor thrombus who underwent surgical treatment at Peking University Third Hospital between January 2014 and December 2021. The patients were divided into floating morphology (107 patients) and filled morphology (122 patients) tumor thrombi groups. Chi-square and Mann-Whitney U tests were used for categorical and continuous variables, respectively. Postoperative complications were evaluated using the Clavien-Dindo surgical complication classification method. RESULTS: Patients with filled morphology tumor thrombus required more surgical techniques than those with floating morphology tumor thrombus, which was reflected in more open surgeries (P < 0.001), more IVC interruptions (P <0.001), lesser use of the delayed occlusion of the proximal inferior vena cava (DOPI) technique (P < 0.001), and a greater need for cut-off of the short hepatic vein (P < 0.001) and liver dissociation (P = 0.001). Filled morphology significantly increased the difficulty of surgery in patients with renal cell carcinoma with tumor thrombus, reflected in longer operation time (P < 0.001), more surgical blood loss (P <0.001), more intra-operative blood transfusion (P < 0.001), and longer postoperative hospital stay (P < 0.001). Filled morphology tumor thrombus also led to more postoperative complications (53% vs. 20%; P < 0.001). CONCLUSION: Compared with floating morphology thrombus, filled morphology thrombus significantly increased the difficulty of surgery in patients with renal cell carcinoma with IVC tumor thrombus.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Cellules tumorales circulantes , Veine cave inférieure , Thrombose veineuse , Humains , Néphrocarcinome/chirurgie , Néphrocarcinome/anatomopathologie , Tumeurs du rein/chirurgie , Tumeurs du rein/anatomopathologie , Veine cave inférieure/anatomopathologie , Veine cave inférieure/chirurgie , Femelle , Mâle , Études rétrospectives , Adulte d'âge moyen , Chine/épidémiologie , Cellules tumorales circulantes/anatomopathologie , Thrombose veineuse/anatomopathologie , Thrombose veineuse/chirurgie , Sujet âgé , Néphrectomie/méthodes , Complications postopératoires/épidémiologie , Adulte
20.
PLoS One ; 19(7): e0306450, 2024.
Article de Anglais | MEDLINE | ID: mdl-39083508

RÉSUMÉ

Metastasis remains the leading cause of cancer deaths worldwide and lung cancer, known for its highly metastatic progression, remains among the most lethal of malignancies. Lung cancer metastasis can selectively spread to multiple different organs, however the genetic and molecular drivers for this process are still poorly understood. Understanding the heterogeneous genomic profile of lung cancer metastases is considered key in identifying therapeutic targets that prevent its spread. Research has identified the key source for metastasis being clusters of cells rather than individual cancer cells. These clusters, known as metastatic cancer cell clusters (MCCCs) have been shown to be 100-fold more tumorigenic than individual cancer cells. Unfortunately, access to these primary drivers of metastases remains difficult and has limited our understanding of their molecular and genomic profiles. Strong evidence in the literature suggests that differentially regulated biological pathways in MCCCs can provide new therapeutic drug targets to help combat cancer metastases. In order to expand research into MCCCs and their role in metastasis, we demonstrate a novel, proof of principle technology, to capture MCCCs directly from patients' whole blood. Our platform can be readily tuned for different solid tumor types by combining a biomimicry-based margination effect coupled with immunoaffinity to isolate MCCCs. Adopting a selective capture approach based on overexpressed CD44 in MCCCs provides a methodology that preferentially isolates them from whole blood. Furthermore, we demonstrate a high capture efficiency of more than 90% when spiking MCCC-like model cell clusters into whole blood. Characterization of the captured MCCCs from lung cancer patients by immunofluorescence staining and genomic analyses, suggests highly differential morphologies and genomic profiles. This study lays the foundation to identify potential drug targets thus unlocking a new area of anti-metastatic therapeutics.


Sujet(s)
Tumeurs du poumon , Métastase tumorale , Cellules tumorales circulantes , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/secondaire , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Étude de validation de principe , Génomique , Femelle , Mâle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE