Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunol ; 24(1): 29-41, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22190576

ABSTRACT

The clinical application of dendritic cells (DC) as adjuvants in immunotherapies such as the cell-based cancer vaccine continues to gain interest. The overall efficacy of this emerging immunotherapy, however, remains low. Studies suggest the stage of maturation and activation of ex vivo-prepared DC immediately prior to patient administration is critical to subsequent DC migration in vivo, which ultimately affects overall vaccine efficacy. While it is possible to generate mature and activated DC ex vivo using various stimulatory cocktails, in the case of cancer patients, the qualitative and quantitative assessment of which DC stimulatory cocktail works most effectively to enhance subsequent DC migration in vivo is difficult. Thus, a non-invasive imaging modality capable of monitoring the real-time migration of DC in long-term studies is required. In this paper, we address whether cellular magnetic resonance imaging (MRI) is sufficiently sensitive to quantitatively detect differences in the migratory abilities of two different DC preparations: untreated (resting) versus ex vivo matured in a mouse model. In order to distinguish our ex vivo-generated DC of interest from surrounding tissues in magnetic resonance (MR) images, DC were labeled in vitro with the superparamagnetic iron oxide (SPIO) nanoparticle FeREX®. Characterization of DC phenotype and function following addition of a cytokine maturation cocktail and the toll-like receptor ligand CpG, both in the presence and in the absence of SPIO, were also carried out. Conventional histological techniques were used to verify the quantitative data obtained from MR images. This study provides important information relevant to tracking the in vivo migration of ex vivo-prepared and stimulated DC.


Subject(s)
Bone Marrow Cells/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Magnetic Resonance Imaging/methods , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Bone Marrow Cells/metabolism , Cancer Vaccines/immunology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Cytokines/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Ferric Compounds/chemistry , Flow Cytometry , Humans , Immunotherapy/methods , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/chemistry , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/metabolism , Oligodeoxyribonucleotides/pharmacology , Ovalbumin/immunology , Ovalbumin/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Reproducibility of Results , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
2.
Cytotherapy ; 13(10): 1234-48, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21923625

ABSTRACT

BACKGROUND AIMS. The use of dendritic cells (DC) as an adjuvant in cell-based immunotherapeutic cancer vaccines is a growing field of interest. A reliable and non-invasive method to track the fate of autologous DC following their administration to patients is required in order to confirm that clinically sufficient numbers are reaching the lymph node (LN). We demonstrate that an immunocompromised mouse model can be used to conduct translational studies employing cellular magnetic resonance imaging (MRI). Such studies can provide clinically relevant information regarding the migration potential of clinical-grade DC used in cancer immunotherapies. METHODS. Human monocyte-derived dendritic cells (mo-DC) were generated from negatively selected monocytes obtained from either healthy donors or cancer patients. DC were labeled with superparamagnetic iron oxide (SPIO) nanoparticles in order to track them in vivo in a CB17scid mouse model using cellular MRI. SPIO did not have any adverse effects on DC phenotype or function, independent of donor type. Cellular MRI readily detected migration of SPIO-loaded DC in CB17scid mice. No differences in migration were observed between DC obtained from healthy donors and those obtained from donors undergoing autologous stem cell transplant for cancer therapy. CONCLUSIONS. Cellular MRI provided semi-quantitative image data that corresponded with data obtained by digital morphometry, validating cellular MRI's potential to assess DC migration in DC-based cancer immunotherapy clinical trials.


Subject(s)
Cancer Vaccines , Cell Movement , Dendritic Cells/metabolism , Immunotherapy, Adoptive , Neoplasms/therapy , Animals , Antigens, Differentiation/metabolism , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/transplantation , Disease Models, Animal , Feasibility Studies , Humans , Magnetic Resonance Imaging/methods , Mice , Mice, SCID , Monocytes/cytology , Neoplasms/immunology , Neoplasms/pathology
3.
Contrast Media Mol Imaging ; 6(4): 314-27, 2011.
Article in English | MEDLINE | ID: mdl-21861291

ABSTRACT

An optimized non-invasive imaging modality capable of tracking and quantifying in vivo DC migration in patients would provide clinicians with valuable information regarding therapeutic DC-based vaccine outcomes. Superparamagnetic iron oxide (SPIO) nanoparticles were used to label bone marrow-derived DC. In vivo DC migration was tracked and quantified non-invasively using cellular magnetic resonance imaging (MRI) in a mouse model. Labelling DC with SPIO reflects the kinetics of DC migration in vivo but appears to reduce overall DC migration, in part due to nanoparticle size. Magnetic separation of SPIO-labelled (SPIO(+)) DC from unlabelled (SPIO(-)) DC prior to injection improves SPIO(+) DC migration to the lymph node. Corresponding MR image data better correlate with the presence of DC in vivo; an improved immunological response is also seen. Cellular MRI is a viable, non-invasive imaging tool that can routinely track DC migration in vivo. Consideration should be given to optimizing MRI contrast agent-labelling of clinical-grade DC in order to accurately correlate DC fate to immunological outcomes in patients.


Subject(s)
Contrast Media , Dendritic Cells/cytology , Magnetic Resonance Imaging/methods , Animals , Cell Movement/physiology , Cells, Cultured , Male , Mice , Mice, Inbred C57BL
4.
Mol Imaging Biol ; 13(4): 679-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20803172

ABSTRACT

PURPOSE: This study seeks to assess the use of labeling with micron-sized iron oxide (MPIO) particles for the detection and quantification of the migration of dendritic cells (DCs) using cellular magnetic resonance imaging (MRI). PROCEDURES: DCs were labeled with red fluorescent MPIO particles for detection by cellular MRI and a green fluorescent membrane dye (PKH67) for histological detection. MPIO-labeled DCs or unlabeled control DCs were injected into mice footpads at two doses (0.1 × 10(6) and 1 × 10(6)). Images were acquired at 3 Tesla before DC injection and 2, 3, and 7 days post-DC injection. RESULTS: Labeling DCs with MPIO particles did not affect viability, but it did alter markers of DC activation and maturation. MRI and fluorescence microscopy allowed for the detection of MPIO-labeled DCs within the draining popliteal nodes after their injection into the footpad. CONCLUSIONS: This paper presents the first report of the successful use of fluorescent MPIO particles to label and track DC migration.


Subject(s)
Cell Movement , Dendritic Cells/cytology , Ferric Compounds/chemistry , Ferric Compounds/metabolism , Magnetic Resonance Imaging/methods , Particle Size , Animals , Apoptosis , Bone Marrow Cells/cytology , Cell Survival , Dendritic Cells/metabolism , Fluorescence , Lymph Nodes/pathology , Male , Mice , Mice, Inbred C57BL , Organic Chemicals/metabolism , Phenotype , Staining and Labeling
5.
J Immunother ; 32(3): 240-51, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19242376

ABSTRACT

Despite recent therapeutic advances, including the introduction of novel cytostatic drugs and therapeutic antibodies, many cancer patients will experience recurrent or metastatic disease. Current treatment options, particularly for those patients with metastatic breast, prostate, or skin cancers, are complex and have limited curative potential. Recent clinical trials, however, have shown that cell-based therapeutic vaccines may be used to generate broad-based, antitumor immune responses. Dendritic cells (DC) have proved to be the most efficacious cellular component for therapeutic vaccines, serving as both the adjuvant and antigen delivery vehicle. At present it is not possible to noninvasively determine the fate of DC-based vaccines after their administration to human subjects. In this study, we demonstrate that in vitro-generated mouse DC can be readily labeled with superparamagnetic iron oxide nanoparticles, Feridex, without altering cell morphology, or their phenotypic and functional maturation. Feridex-labeling enables the detection of DC in vivo after their migration to draining lymph nodes using a 1.5 T clinical magnetic resonance scanner. In addition, we report a semiquantitative approach for analysis of magnetic resonance images and show that the Feridex-induced signal void volume, and fractional signal loss, correlates with the delivery and migration of small numbers of in vitro-generated DC. These findings, together with ongoing preclinical studies, are key to gaining information critical for improving the efficacy of therapeutic vaccines for the treatment cancer, and potentially, chronic infectious diseases.


Subject(s)
Cell Movement/physiology , Dendritic Cells/physiology , Magnetic Resonance Imaging/methods , Animals , Cell Movement/drug effects , Contrast Media/chemistry , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dextrans , Ferrosoferric Oxide , Iron/chemistry , Magnetite Nanoparticles , Mice , Mice, Inbred C57BL , Oxides/chemistry
6.
Virology ; 375(1): 48-58, 2008 May 25.
Article in English | MEDLINE | ID: mdl-18291435

ABSTRACT

A current goal of vaccine development against human immunodeficiency virus (HIV) is to develop a strategy that stimulates long-lasting memory T-cell responses, and provides immediate cytotoxicity in response to viral challenge. We demonstrate that the viral antiapoptotic molecule M11L promotes cellular immune responses to the HIV envelope protein. Coexpression of M11L in vitro inhibits gp140-mediated apoptosis and increases gp140 expression levels. Mice primed with M11L-pHERO DNA, followed by vCP205 boosting, exhibit significantly greater HIV-specific T-cell responses. Moreover, M11L synergizes with CpG motifs to augment anti-HIV responses and stimulates robust expansion of central memory and effector memory CD8(+) T-cells. Inclusion of M11L in a DNA vector increases the magnitude of T-cell responses, and promotes the generation of memory T-cells that provide rapid-responding CTL responses. This vaccine strategy may facilitate the generation of an efficacious vaccine for HIV, and other chronic diseases that require enhanced cell-mediated immunity, including HCV and metastatic cancer.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Vaccines, DNA/immunology , Viral Proteins/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/genetics , Animals , Flow Cytometry , HIV Infections/immunology , HIV Infections/prevention & control , Immunization, Secondary , Immunologic Memory , Male , Mice , Mice, Inbred BALB C , T-Lymphocyte Subsets/immunology , Vaccines, DNA/genetics , Viral Proteins/genetics , env Gene Products, Human Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...