Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
PLoS One ; 19(6): e0304505, 2024.
Article in English | MEDLINE | ID: mdl-38889180

ABSTRACT

The role of macrophages in regulating the tumor microenvironment has spurned the exponential generation of nanoparticle targeting technologies. With the large amount of literature and the speed at which it is generated it is difficult to remain current with the most up-to-date literature. In this study we performed a topic modeling analysis of 854 abstracts of peer-reviewed literature for the most common usages of nanoparticle targeting of tumor associated macrophages (TAMs) in solid tumors. The data spans 20 years of literature, providing a broad perspective of the nanoparticle strategies. Our topic model found 6 distinct topics: Immune and TAMs, Nanoparticles, Imaging, Gene Delivery and Exosomes, Vaccines, and Multi-modal Therapies. We also found distinct nanoparticle usage, tumor types, and therapeutic trends across these topics. Moreover, we established that the topic model could be used to assign new papers into the existing topics, thereby creating a Living Review. This type of "birds-eye-view" analysis provides a useful assessment tool for exploring new and emerging themes within a large field.


Subject(s)
Machine Learning , Nanoparticles , Nanoparticles/chemistry , Humans , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Neoplasms , Tumor Microenvironment , Animals
2.
bioRxiv ; 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37425838

ABSTRACT

Monocytes are members of the mononuclear phagocyte system involved in pathogen clearance and nanoparticle pharmacokinetics. Monocytes play a critical role in the development and progression of cardiovascular disease and, recently, in SARS-CoV-2 pathogenesis. While studies have investigated the effect of nanoparticle modulation on monocyte uptake, their capacity for nanoparticle clearance is poorly studied. In this study, we investigated the impact of ACE2 deficiency, frequently observed in individuals with cardiovascular complications, on monocyte nanoparticle endocytosis. Moreover, we investigated nanoparticle uptake as a function of nanoparticle size, physiological shear stress, and monocyte phenotype. Our Design of Experiment (DOE) analysis found that the THP-1 ACE2 - cells showed a greater preference for 100nm particles under atherosclerotic conditions than THP-1 wild-type cells. Observing how nanoparticles can modulate monocytes in the context of disease can inform precision dosing.

3.
bioRxiv ; 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37425888

ABSTRACT

The role of macrophages in regulating the tumor microenvironment has spurned the exponential generation of nanoparticle targeting technologies. With the large amount of literature and the speed at which it is generated it is difficult to remain current with the most up-to-date literature. In this study we performed a topic modeling analysis of the most common usages of nanoparticle targeting of macrophages in solid tumors. The data spans 20 years of literature, providing an extensive meta-analysis of the nanoparticle strategies. Our topic model found 6 distinct topics: Immune and TAMs, Nanoparticles, Imaging, Gene Delivery and Exosomes, Vaccines, and Multi-modal Therapies. We also found distinct nanoparticle usage, tumor types, and therapeutic trends across these topics. Moreover, we established that the topic model could be used to assign new papers into the existing topics, thereby creating a Living Review. This type of meta-analysis provides a useful assessment tool for aggregating data about a large field.

4.
Front Bioeng Biotechnol ; 11: 1159004, 2023.
Article in English | MEDLINE | ID: mdl-37251577

ABSTRACT

We report on the use of a lab-on-CMOS biosensor platform for quantitatively tracking the proliferation of RAW 264.7 murine Balb/c macrophages. We show that macrophage proliferation correlates linearly with an average capacitance growth factor resulting from capacitance measurements at a plurality of electrodes dispersed in a sensing area of interest. We further show a temporal model that captures the cell number evolution in the area over long periods (e.g., 30 h). The model links the cell numbers and the average capacitance growth factor to describe the observed cell proliferation.

5.
bioRxiv ; 2023 Jan 28.
Article in English | MEDLINE | ID: mdl-36747857

ABSTRACT

We report on the use of a lab-on-CMOS biosensor platform for quantitatively tracking the growth of RAW 264.7 murine Balb/c macrophages. We show that macrophage growth over a wide sensing area correlates linearly with an average capacitance growth factor resulting from capacitance measurements at a plurality of electrodes dispersed in the sensing area. We further show a temporal model that captures the cell evolution in the area of interest over long periods (e.g., 30 hours). The model links the cell numbers and the average capacitance growth factor associated with the sensing area to describe the observed growth kinetics.

6.
Adv Drug Deliv Rev ; 182: 114116, 2022 03.
Article in English | MEDLINE | ID: mdl-35085623

ABSTRACT

Due to the increasing population of individuals with cardiovascular diseases and related comorbidities, there is an increasing need for development of synergistic therapeutics. Monocytes are implicated in a broad spectrum of diseases and can serve as a focal point for therapeutic targeting. This review discusses the role of monocytes in cardiovascular diseases and highlights trends in monocyte targets nanoparticles in three cardiovascular-related diseases: Diabetes, Atherosclerosis, and HIV. Finally, the review offers perspectives on how to develop nanoparticle monocyte targeting strategies that can be beneficial for treating co-morbidities.


Subject(s)
Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Macrophages/metabolism , Monocytes/metabolism , Nanoparticle Drug Delivery System/chemistry , Atherosclerosis/drug therapy , Atherosclerosis/epidemiology , Atherosclerosis/physiopathology , Cardiovascular Diseases/epidemiology , Comorbidity , Diabetes Mellitus/drug therapy , Diabetes Mellitus/epidemiology , Diabetes Mellitus/physiopathology , HIV Infections/drug therapy , HIV Infections/epidemiology , HIV Infections/physiopathology , Humans , Pharmacokinetics , Pharmacology
7.
Nat Rev Mater ; 7(1): 2-4, 2022 Jan.
Article in English | MEDLINE | ID: mdl-37200939

ABSTRACT

Bioengineered platforms, intended to be used in the investigation of human health and disease, often incorporate cells of unknown ancestry or that lack diversity. To develop tools and platforms that benefit the entire human population, we must consider the ancestry of cells and intentionally diversify the cells we use in our designs.

8.
MRS Bull ; 46(9): 847-853, 2021.
Article in English | MEDLINE | ID: mdl-34608355

ABSTRACT

ABSTRACT: From Operation Warp Speed to the lipid mRNA vaccine, the COVID-19 pandemic has been a watershed moment for technological development, production, and implementation. The scale and pace of innovation and global collaboration has likely not been experienced since World War II. This article highlights some of the engineering accomplishments that occurred during the pandemic. We provide a broad overview of the technological achievements in vaccine design, antibody engineering, drug repurposing, and rapid diagnostic testing. We also discuss what the future of these technologies and the future of large-scale collaborations might look like moving forward.

9.
Bio Protoc ; 11(6): e3953, 2021 Mar 20.
Article in English | MEDLINE | ID: mdl-33855115

ABSTRACT

Tumor xenograft models developed by transplanting human tissues or cells into immune-deficient mice are widely used to study human cancer response to drug candidates. However, immune-deficient mice are unfit for investigating the effect of immunotherapeutic agents on the host immune response to cancer (Morgan, 2012). Here, we describe the preparation of an orthotopic, syngeneic model of lung adenocarcinoma (LUAD), a subtype of non-small cell lung cancer (NSCLC), to study the antitumor effect of chemo and immunotherapeutic agents in an immune-competent animal. The tumor model is developed by implanting 344SQ LUAD cells derived from the metastases of KrasG12D; p53R172HΔG genetically engineered mouse model into the left lung of a syngeneic host (Sv/129). The 344SQ LUAD model offers several advantages over other models: 1) The immune-competent host allows for the assessment of the biologic effects of immune-modulating agents; 2) The pathophysiological features of the human disease are preserved due to the orthotopic approach; 3) Predisposition of the tumor to metastasize facilitates the study of therapeutic effects on primary tumor as well as the metastases ( Chen et al., 2014 ). Furthermore, we also describe a treatment strategy based on Poly(2-oxazoline) micelles that has been shown to be effective in this difficult-to-treat tumor model ( Vinod et al., 2020b ).

10.
Bio Protoc ; 11(6): e3959, 2021 Mar 20.
Article in English | MEDLINE | ID: mdl-33855119

ABSTRACT

Many new drug development candidates are highly lipophilic compounds with low water solubility. This constitutes a formidable challenge for the use of such compounds for cancer therapy, where high doses and intravenous injections are needed ( Di et al., 2012 ). Here, we present a poly(2-oxazoline) polymer (POx)-based nanoformulation strategy to solubilize and deliver hydrophobic drugs. POx micelles are prepared by a simple thin-film hydration method. In this method, the drug and polymer are dissolved in a common solvent and allowed to mix, following which the solvent is evaporated using mild heating conditions to form a thin film. The micelles form spontaneously upon hydration with saline. POx nanoformulation of hydrophobic drugs is unique in that it has a high drug loading capacity, which is superior to micelles of conventional surfactants. Moreover, multiple active pharmaceutical ingredients (APIs) can be included within the same POx micelle, thereby enabling the codelivery of binary as well as ternary drug combinations ( Han et al., 2012 ; He et al., 2016 ).

11.
Front Immunol ; 12: 566299, 2021.
Article in English | MEDLINE | ID: mdl-33732229

ABSTRACT

Extracellular vesicles (EVs) are important players in autoimmune diseases, both in disease pathogenesis and as potential treatments. EVs can transport autoimmune triggers throughout the body, facilitating the process of antigen presentation. Understanding the link between cellular stress and EV biogenesis and intercellular trafficking will advance our understanding of autoimmune diseases. In addition, EVs can also be effective treatments for autoimmune diseases. The diversity of cell types that produce EVs leads to a wide range of molecules to be present in EVs, and thus EVs have a wide range of physiological effects. EVs derived from dendritic cells or mesenchymal stem cells have been shown to reduce inflammation. Since many autoimmune treatments are focused only on symptom management, EVs present a promising avenue for potential treatments. This review looks at the different roles EVs can play in autoimmune diseases, from disease pathology to diagnosis and treatment. We also overview various methodologies in isolating or generating EVs and look to the future for possible applications of EVs in autoimmune diseases.


Subject(s)
Antigen Presentation/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Extracellular Vesicles/immunology , Animals , Autoantigens/immunology , Autoantigens/metabolism , Autoimmune Diseases/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Extracellular Vesicles/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Stress, Physiological/immunology
12.
Sci Adv ; 6(25): eaba5542, 2020 06.
Article in English | MEDLINE | ID: mdl-32596460

ABSTRACT

About 40% of patients with non-small cell lung cancer (NSCLC) have stage IV cancer at the time of diagnosis. The only viable treatment options for metastatic disease are systemic chemotherapy and immunotherapy. Nonetheless, chemoresistance remains a major cause of chemotherapy failure. New immunotherapeutic modalities such as anti-PD-1 immune checkpoint blockade have shown promise; however, response to such strategies is highly variable across patients. Here, we show that our unique poly(2-oxazoline)-based nanomicellar formulation (PM) of Resiquimod, an imidazoquinoline Toll-like receptor (TLR) 7/8 agonist, had a superior tumor inhibitory effect in a metastatic model of lung adenocarcinoma, relative to anti-PD-1 therapy or platinum-based chemotherapy. Investigation of the in vivo immune status following Resiquimod PM treatment showed that Resiquimod-based stimulation of antigen-presenting cells in the tumor microenvironment resulted in the mobilization of an antitumor CD8+ immune response. Our study demonstrates the promise of poly(2-oxazoline)-formulated Resiquimod for treating metastatic NSCLC.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adenocarcinoma of Lung/drug therapy , Adjuvants, Immunologic , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Oxazoles , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Tumor Microenvironment
13.
Adv Sci (Weinh) ; 6(21): 1900582, 2019 Nov 06.
Article in English | MEDLINE | ID: mdl-31728272

ABSTRACT

Delivery of nucleic acids into solid tumor environments remains a pressing challenge. This study examines the ability of macrophages to horizontally transfer small interfering RNA (siRNA) lipoplexes to cancer cells. Macrophages are a natural candidate for a drug carrier because of their ability to accumulate at high densities into many cancer types, including, breast, prostate, brain, and colon cancer. Here, it is demonstrated that macrophages can horizontally transfer siRNA to cancer cells during in vitro coculture. The amount of transfer can be dosed depending on the amount of siRNA loaded and total number of macrophages delivered. Macrophages loaded with calcium integrin binding protein-1 (CIB1)-siRNA result in decreased tumorsphere growth and decreased mRNA expression of CIB1 and KI67 in MDA-MB-468 human breast cancer cells. Adoptive transfer of macrophages transfected with CIB1-siRNA localizes to the orthotopic MDA-MB-468 tumor. Furthermore, it is reported that macrophage activation can modulate this transfer process as well as intracellular trafficking protein Rab27a. As macrophages are heavily involved in tumor progression, understanding how to use macrophages for drug delivery can substantially benefit the treatment of tumors.

15.
Oncotarget ; 8(3): 3826-3839, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27270652

ABSTRACT

TGFß has both tumor suppressive and tumor promoting effects in colon cancer. Also, TGFß can affect the extent and composition of inflammatory cells present in tumors, contextually promoting and inhibiting inflammation. While colon tumors display intratumoral inflammation, the contributions of TGFß to this process are poorly understood. In human patients, we found that epithelial loss of TGFß signaling was associated with increased inflammatory burden; yet overexpression of TGFß was also associated with increased inflammation. These findings were recapitulated in mutant APC models of murine tumorigenesis, where epithelial truncation of TGFBR2 led to lethal inflammatory disease and invasive colon cancer, mediated by IL8 and TGFß1. Interestingly, mutant APC mice with global suppression of TGFß signals displayed an intermediate phenotype, presenting with an overall increase in IL8-mediated inflammation and accelerated tumor formation, yet with a longer latency to the onset of disease observed in mice with epithelial TGFBR-deficiency. These results suggest that the loss of TGFß signaling, particularly in colon epithelial cells, elicits a strong inflammatory response and promotes tumor progression. This implies that treating colon cancer patients with TGFß inhibitors may result in a worse outcome by enhancing inflammatory responses.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Colonic Neoplasms/pathology , Cytokines/metabolism , Transforming Growth Factor beta/metabolism , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms, Experimental , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad4 Protein/metabolism
16.
Cancer Res ; 76(9): 2525-39, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26980767

ABSTRACT

In early pancreatic carcinogenesis, TGFß acts as a tumor suppressor due to its growth-inhibitory effects in epithelial cells. However, in advanced disease, TGFß appears to promote tumor progression. Therefore, to better understand the contributions of TGFß signaling to pancreatic carcinogenesis, we generated mouse models of pancreatic cancer with either epithelial or systemic TGFBR deficiency. We found that epithelial suppression of TGFß signals facilitated pancreatic tumorigenesis, whereas global loss of TGFß signaling protected against tumor development via inhibition of tumor-associated fibrosis, stromal TGFß1 production, and the resultant restoration of antitumor immune function. Similarly, TGFBR-deficient T cells resisted TGFß-induced inactivation ex vivo, and adoptive transfer of TGFBR-deficient CD8(+) T cells led to enhanced infiltration and granzyme B-mediated destruction of developing tumors. These findings paralleled our observations in human patients, where TGFß expression correlated with increased fibrosis and associated negatively with expression of granzyme B. Collectively, our findings suggest that, despite opposing the proliferation of some epithelial cells, TGFß may promote pancreatic cancer development by affecting stromal and hematopoietic cell function. Therefore, the use of TGFBR inhibition to target components of the tumor microenvironment warrants consideration as a potential therapy for pancreatic cancer, particularly in patients who have already lost tumor-suppressive TGFß signals in the epithelium. Cancer Res; 76(9); 2525-39. ©2016 AACR.


Subject(s)
Carcinogenesis/metabolism , Pancreatic Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Tumor Escape/physiology , Tumor Microenvironment/physiology , Adoptive Transfer , Animals , Blotting, Western , Carcinogenesis/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fibrosis/metabolism , Fibrosis/pathology , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Mutant Strains , Signal Transduction/physiology
17.
J Control Release ; 223: 215-223, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26732555

ABSTRACT

Prostate cancer, once it has progressed from its local to metastatic form, is a disease with poor prognosis and limited treatment options. Here we demonstrate an approach using nanoscale liposomes conjugated with E-selectin adhesion protein and Apo2L/TRAIL (TNF-related apoptosis-inducing ligand) apoptosis ligand that attach to the surface of leukocytes and rapidly clear viable cancer cells from circulating blood in the living mouse. For the first time, it is shown that such an approach can be used to prevent the spontaneous formation and growth of metastatic tumors in an orthotopic xenograft model of prostate cancer, by greatly reducing the number of circulating tumor cells. We conclude that the use of circulating leukocytes as a carrier for the anti-cancer protein TRAIL could be an effective tool to directly target circulating tumor cells for the prevention of prostate cancer metastasis, and potentially other cancers that spread through the bloodstream.


Subject(s)
E-Selectin/administration & dosage , Leukocytes , Neoplastic Cells, Circulating/drug effects , Prostatic Neoplasms/drug therapy , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Animals , E-Selectin/therapeutic use , Humans , Leukocytes/metabolism , Liposomes , Male , Mice, Transgenic , Neoplasm Metastasis/prevention & control , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Xenograft Model Antitumor Assays
18.
BMC Cancer ; 15: 206, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25884197

ABSTRACT

BACKGROUND: Circulating biomarkers are urgently needed in hepatocellular carcinoma (HCC). The aims of this study were to determine the feasibility of detecting and isolating circulating tumor cells (CTCs) in HCC patients using enrichment for epithelial cell adhesion molecule (EpCAM) expression, to examine their prognostic value, and to explore CTC-based DNA sequencing in metastatic HCC patients compared to a control cohort with non-malignant liver diseases (NMLD). METHODS: Whole blood was obtained from patients with metastatic HCC or NMLD. CTCs were enumerated by CellSearch then purified by immunomagnetic EpCAM enrichment and fluorescence-activated cell sorting. Targeted ion semiconductor sequencing was performed on whole genome-amplified DNA from CTCs, tumor specimens, and peripheral blood mononuclear cells (PBMC) when available. RESULTS: Twenty HCC and 10 NMLD patients enrolled. CTCs ≥ 2/7.5 mL were detected in 7/20 (35%, 95% confidence interval: 12%, 60%) HCC and 0/9 eligible NMLD (p = 0.04). CTCs ≥ 1/7.5 mL was associated with alpha-fetoprotein ≥ 400 ng/mL (p = 0.008) and vascular invasion (p = 0.009). Sequencing of CTC DNA identified characteristic HCC mutations. The proportion with ≥ 100x coverage depth was lower in CTCs (43%) than tumor or PBMC (87%) (p < 0.025). Low frequency variants were higher in CTCs (p < 0.001). CONCLUSIONS: CTCs are detectable by EpCAM enrichment in metastatic HCC, without confounding false positive background from NMLD. CTC detection was associated with poor prognostic factors. Sequencing of CTC DNA identified known HCC mutations but more low-frequency variants and lower coverage depth than FFPE or PBMC.


Subject(s)
Antigens, Neoplasm/genetics , Carcinoma, Hepatocellular/genetics , Cell Adhesion Molecules/genetics , Liver Neoplasms/genetics , Neoplastic Cells, Circulating , Aged , Aged, 80 and over , Antigens, Neoplasm/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules/blood , Epithelial Cell Adhesion Molecule , Epithelial-Mesenchymal Transition/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Kaplan-Meier Estimate , Liver Diseases/blood , Liver Diseases/genetics , Liver Diseases/pathology , Liver Neoplasms/blood , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Polymorphism, Single Nucleotide , Prognosis
19.
PLoS One ; 9(11): e112802, 2014.
Article in English | MEDLINE | ID: mdl-25426709

ABSTRACT

Recombinant subunit vaccine engineering increasingly focuses on the development of more effective delivery platforms. However, current recombinant vaccines fail to sufficiently stimulate protective adaptive immunity against a wide range of pathogens while remaining a cost effective solution to global health challenges. Taking an unorthodox approach to this fundamental immunological challenge, we isolated the TLR-targeting capability of the probiotic E. coli Nissle 1917 bacteria (EcN) by engineering bionanoparticlate antigen carriers derived from EcN outer membrane vesicles (OMVs). Exogenous model antigens expressed by these modified bacteria as protein fusions with the bacterial enterotoxin ClyA resulted in their display on the surface of the carrier OMVs. Vaccination with the engineered EcN OMVs in a BALB/c mouse model, and subsequent mechanism of action analysis, established the EcN OMV's ability to induce self-adjuvanted robust and protective humoral and T(H)1-biased cellular immunity to model antigens. This finding appears to be strain-dependent, as OMV antigen carriers similarly engineered from a standard K12 E. coli strain derivative failed to generate a comparably robust antigen-specific TH1 bias. The results demonstrate that unlike traditional subunit vaccines, these biomolecularly engineered "pathogen-like particles" derived from traditionally overlooked, naturally potent immunomodulators have the potential to effectively couple recombinant antigens with meaningful immunity in a broadly applicable fashion.


Subject(s)
Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Escherichia coli/immunology , Th1 Cells/immunology , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Cell Membrane/chemistry , Cell Membrane/immunology , Escherichia coli/chemistry , Escherichia coli Proteins/administration & dosage , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Female , Gene Expression , Hemolysin Proteins/administration & dosage , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunization , Mice , Mice, Inbred BALB C , Probiotics/chemistry , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Species Specificity , Th1 Cells/cytology , Vaccines, Subunit , Vaccines, Synthetic
20.
Proc Natl Acad Sci U S A ; 111(3): 930-5, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395803

ABSTRACT

Metastasis through the bloodstream contributes to poor prognosis in many types of cancer. Mounting evidence implicates selectin-based adhesive interactions between cancer cells and the blood vessel wall as facilitating this process, in a manner similar to leukocyte trafficking during inflammation. Here, we describe a unique approach to target and kill colon and prostate cancer cells in the blood that causes circulating leukocytes to present the cancer-specific TNF-related apoptosis inducing ligand (TRAIL) on their surface along with E-selectin adhesion receptor. This approach, demonstrated in vitro with human blood and also in mice, mimics the cytotoxic activity of natural killer cells and increases the surface area available for delivery of the receptor-mediated signal. The resulting "unnatural killer cells" hold promise as an effective means to neutralize circulating tumor cells that enter blood with the potential to form new metastases.


Subject(s)
Apoptosis , Leukocytes/metabolism , Neoplastic Cells, Circulating/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Survival , E-Selectin/metabolism , Endothelial Cells/cytology , Female , Humans , Leukocytes/cytology , Leukocytes, Mononuclear/cytology , Liposomes/chemistry , Liposomes/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplastic Cells, Circulating/pathology , Serum Albumin/metabolism , Shear Strength , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL
...