Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Lancet Oncol ; 16(13): e498-509, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26433823

ABSTRACT

Checkpoint blockade immunotherapy has received mainstream attention as a result of striking and durable clinical responses in some patients with metastatic disease and a reasonable response rate in many tumour types. The activity of checkpoint blockade immunotherapy is not restricted to melanoma or lung cancer, and additional indications are expected in the future, with responses already reported in renal cancer, bladder cancer, and Hodgkin's lymphoma among many others. Additionally, the interactions between radiation and the immune system have been investigated, with several studies describing the synergistic effects on local and distant tumour control when radiation therapy is combined with immunotherapy. Clinical enthusiasm for this approach is strengthened by the many ongoing trials combining immunotherapy with definitive and palliative radiation. Herein, we discuss the biological and mechanistic rationale behind combining radiation with checkpoint blockade immunotherapy, with a focus on the preclinical data supporting this potentially synergistic combination. We explore potential hypotheses and important considerations for clinical trial designs. Finally, we reintroduce the notion of radiosensitising immunotherapy, akin to radiosensitising chemotherapy, as a potential definitive therapeutic modality.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Chemoradiotherapy/methods , Immunotherapy/methods , Neoplasms/therapy , Radiation Tolerance , Radiation-Sensitizing Agents/therapeutic use , Signal Transduction/drug effects , Signal Transduction/radiation effects , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/radiation effects , Dendritic Cells/diagnostic imaging , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/radiation effects , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/pathology , Radiation Dosage , Radiography , Treatment Outcome
2.
Ultrasound Med Biol ; 40(9): 2125-33, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25023114

ABSTRACT

Sonodynamic therapy (SDT) uses a combination of sonosensitizing drugs and low-intensity therapeutic ultrasound to cause apoptosis and autophagy of tumor cells. However, its effects on the tumor microenvironment, especially on the immune state, remain unknown. In this study, we investigated the transformation of macrophages and dendritic cells (DCs) in the tumor microenvironment during 5-aminolevulinic acid (5-ALA)-mediated SDT in mice transplanted with B16F10 melanomas. Tumor growth and mouse weight were measured. Hematoxylin-eosin staining was used to evaluate tumor morphology to quantify the anti-tumor efficacy of 5-ALA-mediated SDT. We investigated anti-tumor immunity in the tumor microenvironment by immunocytochemical staining of CD68, CD163, CD80, CD86, tumor necrosis factor α (TNF-α), interleukin 10 (IL-10) and interferon γ (IFN-γ). Tumor growth was restrained by 5-ALA-mediated SDT in B16F10 melanoma-bearing mice. CD68 levels increased and CD163 decreased, indicating that M2 macrophages were converted to the M1 phenotype in the tumor. The increase in CD80 and CD86 showed that DCs in the tumor microenvironment tend to mature after SDT treatment. The cytokines INF-γ, TNF-α and IL-10 significantly increased in SDT. Application of low-intensity therapeutic ultrasound alone also led to similar trends in our study, but combined treatment with 5-ALA yielded a change. The original stabilized immune state in the tumor microenvironment can be interrupted by low-intensity therapeutic ultrasound combined with 5-ALA, which enhanced the pro-inflammatory response and reversed the passive properties of macrophages and dendritic cells.


Subject(s)
Aminolevulinic Acid/therapeutic use , Dendritic Cells/diagnostic imaging , Macrophages/diagnostic imaging , Melanoma/therapy , Ultrasonic Therapy/methods , Animals , Cell Line, Tumor , Cell Survival/drug effects , Combined Modality Therapy/methods , Dendritic Cells/drug effects , Disease Models, Animal , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Photosensitizing Agents/therapeutic use , Ultrasonography
3.
Exp Biol Med (Maywood) ; 239(1): 13-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24227632

ABSTRACT

Dendritic cells (DCs) play an important role in atherosclerosis plaque formation, but the mechanism has not been elucidated clearly. This study is designed to establish a method for tracing DCs in vivo facilitating the investigation of the DCs' specific roles in atherosclerosis. Rabbit DCs labeled by different concentrations of ultra small superparamagnetic iron oxide (USPIO) were injected into atherosclerosis rabbit model and traced with magnetic resonance imaging (MRI). Results showed that USPIO labeling nearly have no cytotoxicity to DCs in low concentrations (<500 µg/mL) but induced some decrease of cell viability at high concentrations (>500 µg/mL). Moreover, USPIO labeling, from 200 to 2000 µg/mL, caused a dose-dependent decrease of the mitochondrial membrane potential in DCs. The high labeling concentration (2000 µg/mL) triggered necrosis instead of apoptosis in DCs. By T2WI and fs T2WI sequence imaging comparison, DCs were found to exist in rabbit abdominal artery plaques after 24 h of transplantation and in spleen after one week detected by Prussian blue staining of tissue sections. We concluded that about 200 µg/mL USPIO is ideal to effectively label DCs for MRI tracing in vivo without a threat to cell viability. Combining USPIO labeling and MRI to track the movement of injected DCs in vivo is a feasible method.


Subject(s)
Dendritic Cells/diagnostic imaging , Ferric Compounds/pharmacology , Magnetic Resonance Imaging , Magnetite Nanoparticles , Animals , Cell Survival/drug effects , Dendritic Cells/metabolism , Dose-Response Relationship, Drug , Ferric Compounds/adverse effects , Ferric Compounds/chemistry , Membrane Potential, Mitochondrial/drug effects , Necrosis/chemically induced , Necrosis/diagnostic imaging , Necrosis/metabolism , Rabbits , Radiography , Spleen/diagnostic imaging
4.
Mol Imaging Biol ; 14(2): 183-96, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21598093

ABSTRACT

PURPOSE: The authors present a protocol for the in vivo evaluation, using different imaging techniques, of lymph node (LN) homing of tumor-specific dendritic cells (DCs) in a murine breast cancer model. PROCEDURES: Bone marrow DCs were labeled with paramagnetic nanoparticles (MNPs) or (111)In-oxine. Antigen loading was performed using tumor lysate. Mature DCs were injected into the footpads of transgenic tumor-bearing mice (MMTV-Ras) and DC migration was tracked by magnetic resonance imaging (MRI) and single-photon emission computed tomography (SPECT). Ex vivo analyses were performed to validate the imaging data. RESULTS: DC labeling, both with MNPs and with (111)In-oxine, did not affect DC phenotype or functionality. MRI and SPECT allowed the detection of iron and (111)In in both axillary and popliteal LNs. Immunohistochemistry and γ-counting revealed the presence of DCs in LNs. CONCLUSIONS: MRI and SPECT imaging, by allowing in vivo dynamic monitoring of DC migration, could further the development and optimization of efficient anti-cancer vaccines.


Subject(s)
Cell Movement , Dendritic Cells/pathology , Diagnostic Imaging/methods , Indium Radioisotopes , Lymph Nodes/diagnostic imaging , Magnetite Nanoparticles , Mammary Neoplasms, Animal/diagnostic imaging , Animals , Cell Separation , Cells, Cultured , Dendritic Cells/diagnostic imaging , Dendritic Cells/ultrastructure , Disease Models, Animal , Female , Genes, ras/genetics , Immunohistochemistry , Lymph Nodes/pathology , Magnetic Resonance Imaging , Magnetite Nanoparticles/ultrastructure , Mammary Neoplasms, Animal/pathology , Mammary Tumor Virus, Mouse/metabolism , Mice , Mice, Transgenic , Phenotype , Staining and Labeling , Tomography, Emission-Computed, Single-Photon
5.
Int J Radiat Oncol Biol Phys ; 82(2): 924-32, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-21398051

ABSTRACT

PURPOSE: The goal of this study was to determine the effect of combination of intratumoral administration of dendritic cells (DC) and fractionated external beam radiation (EBRT) on tumor-specific immune responses in patients with soft-tissue sarcoma (STS). METHODS AND MATERIAL: Seventeen patients with large (>5 cm) high-grade STS were enrolled in the study. They were treated in the neoadjuvant setting with 5,040 cGy of EBRT, split into 28 fractions and delivered 5 days per week, combined with intratumoral injection of 10(7) DCs followed by complete resection. DCs were injected on the second, third, and fourth Friday of the treatment cycle. Clinical evaluation and immunological assessments were performed. RESULTS: The treatment was well tolerated. No patient had tumor-specific immune responses before combined EBRT/DC therapy; 9 patients (52.9%) developed tumor-specific immune responses, which lasted from 11 to 42 weeks. Twelve of 17 patients (70.6%) were progression free after 1 year. Treatment caused a dramatic accumulation of T cells in the tumor. The presence of CD4(+) T cells in the tumor positively correlated with tumor-specific immune responses that developed following combined therapy. Accumulation of myeloid-derived suppressor cells but not regulatory T cells negatively correlated with the development of tumor-specific immune responses. Experiments with (111)In labeled DCs demonstrated that these antigen presenting cells need at least 48 h to start migrating from tumor site. CONCLUSIONS: Combination of intratumoral DC administration with EBRT was safe and resulted in induction of antitumor immune responses. This suggests that this therapy is promising and needs further testing in clinical trials design to assess clinical efficacy.


Subject(s)
Dendritic Cells/transplantation , Sarcoma/therapy , Soft Tissue Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Cell Movement , Combined Modality Therapy/methods , Dendritic Cells/diagnostic imaging , Dendritic Cells/physiology , Dose Fractionation, Radiation , Female , Humans , Immunity, Humoral/immunology , Indium Radioisotopes , Inhibitor of Apoptosis Proteins/immunology , Injections, Intralesional , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Myeloid Cells/immunology , Neoadjuvant Therapy/methods , Neoplasm Proteins/immunology , Radionuclide Imaging , Sarcoma/immunology , Sarcoma/pathology , Sarcoma/radiotherapy , Soft Tissue Neoplasms/immunology , Soft Tissue Neoplasms/pathology , Soft Tissue Neoplasms/radiotherapy , Survivin , T-Lymphocytes/immunology , Time Factors
6.
Adv Drug Deliv Rev ; 62(11): 1080-93, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20800081

ABSTRACT

Cellular therapy promises to revolutionize medicine, by restoring tissue and organ function, and combating key disorders including cancer. As with all major developments, new tools must be introduced to allow optimization. For cell therapy, the key tool is in vivo imaging for real time assessment of parameters such as cell localization, numbers and viability. Such data is critical to modulate and tailor the therapy for each patient. In this review, we discuss recent work in the field of imaging cell therapies in the clinic, including preclinical work where clinical examples are not yet available. Clinical trials in which transferred cells were imaged using magnetic resonance imaging (MRI), nuclear scintigraphy, single photon emission computed tomography (SPECT), and positron emission tomography (PET) are evaluated from an imaging perspective. Preclinical cell tracking studies that focus on fluorescence and bioluminescence imaging are excluded, as these modalities are generally not applicable to clinical cell tracking. In this review, we assess the advantages and drawbacks of the various imaging techniques available, focusing on immune cells, particularly dendritic cells. Both strategies of prelabeling cells before transplant and the use of an injectable label to target cells in situ are covered. Finally, we discuss future developments, including the emergence of multimodal imaging technology for cell tracking from the preclinical to the clinical realm.


Subject(s)
Cell Tracking/methods , Cell Transplantation/diagnostic imaging , Cell Transplantation/methods , Diagnostic Imaging/methods , Animals , Dendritic Cells/diagnostic imaging , Dendritic Cells/transplantation , Humans , Radionuclide Imaging , T-Lymphocytes/diagnostic imaging , T-Lymphocytes/transplantation
7.
Pediatr Radiol ; 40 Suppl 1: S40-2, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20552185

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm (also referred to as CD4+/CD56+ hematodermic neoplasm) is a rare hematological malignancy typically seen in older adults. The disease presents with nonspecific cutaneous lesions and advances toward a fatal leukemic phase despite an often favorable initial response to chemotherapy. Fewer than 200 cases have been documented, and it is exceedingly rare in children. We report a case of histopathologically proven blastic plasmacytoid dendritic cell neoplasm arising in an otherwise healthy and asymptomatic 8-year-old boy who noticed a painless mass within the subcutaneous tissues below the left calf. In addition to contrast-enhanced MRI, the imaging work-up included PET/CT, which showed metastatic involvement within left inguinal and retroperitoneal lymph nodes. The case is notable both for the young age of the patient and for the absence of characteristic cutaneous manifestations of this disease.


Subject(s)
Dendritic Cells/diagnostic imaging , Dendritic Cells/pathology , Diagnostic Imaging/methods , Plasmacytoma/diagnosis , Soft Tissue Neoplasms/diagnosis , Child , Diagnosis, Differential , Humans , Leg/diagnostic imaging , Leg/pathology , Male , Radionuclide Imaging
9.
J Gene Med ; 8(10): 1243-50, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16892456

ABSTRACT

Recombinant DNA vaccines are able to induce strong CD8+ T cell mediated immunity and have become increasingly attractive for the prevention and treatment of infectious diseases and cancer. Dendritic cells (DC), which critically control cellular immune responses, have been transduced with antigen ex vivo and used as 'nature's adjuvant' to enhance vaccine efficacy. The impact of the application route on the in vivo distribution of antigen and the stimulation of CD8+ T cells have been subjects of considerable debate. Here we report the construction of vectors expressing a fusion protein between EGFP, the H2-K(b)-binding peptide OVA(aa257-264) and green click beetle luciferase as a model antigen which allows for simultaneous quantitative assessment of antigen expression using fluorescence and bioluminescence imaging in correlation with CD8+ T cell stimulation in vivo. We applied this construct to evaluate DNA vaccination with recombinant adenoviral vectors, assess the impact of using cultured DC for vaccine delivery and investigate different application routes. Antigen expression was non-invasively followed in vivo by visualizing bioluminescence with an ultrasensitive CCD camera. CD8+ T cell stimulation was detected with H2-K(b)-OVA(aa257-264) tetramers. We found that intravenous injection of adenovirus-transduced DC stimulated the strongest OVA(aa257-264)-specific cytotoxic T-lymphocyte (CTL) responses although it delivered two orders of magnitude less antigen in vivo when compared to direct injection of recombinant adenovirus. We believe that our experimental approach has the potential to facilitate translational development of improved genetic immunization strategies targeting DC directly in vivo.


Subject(s)
Adenoviridae/genetics , Dendritic Cells/transplantation , Microscopy, Fluorescence/methods , Vaccines, DNA/administration & dosage , Animals , Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Cloning, Molecular , Dendritic Cells/diagnostic imaging , Dendritic Cells/immunology , Diagnostic Imaging , Drug Administration Routes , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Luciferases/genetics , Luciferases/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Radiography , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/metabolism
10.
Immunobiology ; 211(6-8): 587-97, 2006.
Article in English | MEDLINE | ID: mdl-16920497

ABSTRACT

Dendritic cells (DCs) play important roles in the initiation of adaptive immune responses. The transport of antigen from the infection site to the draining lymph node by DCs is a crucial component in this process. Accordingly, immunotherapeutic applications of in vitro-generated DCs require reliable methods experimentally in mice and clinically in patients to monitor the efficiency of their successful lymph node homing after injection. Recent developments of new methods to follow DC migration by non-invasive imaging modalities such as scintigraphy, PET, MRI, or bioluminescence imaging, have gained attraction because of their potential clinical applicability. The current state of the literature and a comparative evaluation of the methods are reported in this review.


Subject(s)
Cell Movement/physiology , Dendritic Cells/physiology , Diagnostic Imaging , Animals , Dendritic Cells/diagnostic imaging , Humans , Magnetic Resonance Imaging , Positron-Emission Tomography
11.
Nucl Med Biol ; 33(4): 453-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16720236

ABSTRACT

UNLABELLED: Dendritic cells (DC) are professional antigen-presenting cells capable of inducing potent immune responses. In our ongoing clinical trials, human leukocyte antigen (HLA)-A2.1+ melanoma patients are vaccinated with mature DC, presenting tumor-derived peptides in major histocompatibility complexes (MHC) to naive T cells. Previously, we have shown that both intradermally and intranodally injected (111)In-labeled mature DC migrate to draining lymph nodes. However, little is known about the fate of the MHC-peptide complex after injection of these peptide-loaded DC. The aim of the present study was to develop radiolabeled, tumor-derived peptides to monitor their binding to MHC Class I. METHODS: The HLA-A2.1 binding peptide gp100:154-162mod (gp100:154m) was conjugated with diethylenetriamine pentaacetic acid (DTPA) either at the N-terminus (alpha-DTPA-gp100:154m) or at the epsilon amino group of the Lys(154) residue (epsilon-DTPA-gp100:154m) and labeled with (111)In. RESULTS: The maximum specific activity for both peptides was 13 GBq/micromol. The IC50 of the alpha-[(111)In]DTPA-gp100:154m peptide was >75 microM. The IC50 of the (111)In-labeled epsilon-DTPA-gp100:154m was 3 microM, similar to the unconjugated peptide. MHC binding studies showed specific binding of the epsilon-[(111)In]DTPA-gp100:154m peptide to the JY cells at 4 degrees C. Interestingly, no specific binding was observed for the alpha-[(111)In]DTPA-gp100:154m peptide. In contrast to the alpha-[(111)In]DTPA-gp100:154m peptide, the epsilon-[(111)In]DTPA-gp100:154m peptide was recognized by cytotoxic T cells. CONCLUSION: When DTPA was conjugated to the epsilon NH2 group of the Lys(154) residue, MHC binding of the peptide was preserved and could still be recognized by cytotoxic T cells. These studies allow the noninvasive determination of the behavior of MHC-peptide complexes on DC in vivo.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/transplantation , HLA-A2 Antigen/immunology , Indium Radioisotopes , Peptides/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Binding Sites , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Dendritic Cells/diagnostic imaging , Dendritic Cells/immunology , Humans , Indium Radioisotopes/immunology , Protein Binding , Radionuclide Imaging , Radiopharmaceuticals/immunology
12.
Histopathology ; 43(5): 491-4, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14636276

ABSTRACT

AIMS: Tumours of dendritic/accessory cell origin are rare neoplasms arising in lymph nodes. Among these, tumours derived from cytokeratin-positive interstitial reticulum cells (CIRCs), a subset of fibroblastic reticulum cells, are reported even less frequently. The International Lymphoma Study Group (ILSG) has recently proposed a classification for tumours of histiocytes and accessory dendritic cells in which CIRC tumours are not included. We report a case of a CIRC tumour arising in a submandibular lymph node of a 66-year-old male. METHODS AND RESULTS: The neoplasm was composed of spindle cells with elongated or round nuclei, prominent nucleoli and abundant cytoplasm. These cells were arranged in a diffuse fascicular and vaguely whorled pattern. The tumour cells stained diffusely for S100, vimentin, desmin, lysozyme, and focally for CD68 and cytokeratins 7, 8, 18, CK-AE1 and CK-pool. Electron microscopy was performed for further evaluation on samples taken from the paraffin block; this revealed cytoplasmic projections and rudimentary cell junctions. CONCLUSIONS: Histopathologist should be aware of the existence of tumours deriving from CIRCs, as these cases may be misdiagnosed as metastatic carcinoma. Careful clinical and pathological evaluation is necessary to exclude this possibility.


Subject(s)
Dendritic Cells/pathology , Keratins/metabolism , Lymph Nodes/pathology , Lymphoma/pathology , Submandibular Gland Neoplasms/pathology , Aged , Dendritic Cells/diagnostic imaging , Dendritic Cells/metabolism , Diagnosis, Differential , Humans , Lymph Nodes/metabolism , Lymph Nodes/ultrastructure , Lymphoma/classification , Lymphoma/metabolism , Male , Microscopy, Electron , Submandibular Gland Neoplasms/classification , Submandibular Gland Neoplasms/metabolism , Ultrasonography
13.
Eur J Nucl Med Mol Imaging ; 30(3): 440-7, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12722741

ABSTRACT

In cancer vaccination trials, antigen-loaded dendritic cells (DCs) are usually injected intradermally and are expected to rapidly move to a regional lymph node where antigen presentation should occur. In this study we investigated the influence of indium-111 oxine (111In) and technetium-99m hexamethylpropylene amine oxime (99mTc-HMPAO) labelling on the motility and actin content of antigen-loaded DCs in parallel with in vivo migration in humans. Human autologous monocyte-derived DCs loaded with a tumour antigen were labelled with 111In (0.11, 0.37 or 0.74 MBq/10(7) DCs) or 99mTc-HMPAO (18.5 or 185 MBq/10(7) DCs). 111In labelling was much more stable than 99mTc-HMPAO labelling. Quantitative videomicroscopy showed that the mean distance of displacement of DCs increased in accordance with the 111In activity used for labelling. Monomeric (G) and filamentous (F) actin content of DCs evaluated by quantitative immunofluorescence demonstrated that the ratio of filamentous to globular actin content in labelled DCs increased significantly in accordance with the activity used for labelling with both tracers. Twelve patients enrolled in a phase I/II vaccination trial received injections of 10(7) antigen-loaded DCs labelled with either 0.74 MBq of 111In (group A, n=6/12) or 18.5 MBq of 99mTc-HMPAO (group B, n=6/12) in the proximal part of the legs, one intradermally on one side, one subcutaneously on the opposite side. In three of the six patients of each group, antigen-loaded DCs were incubated with monophosphoryl lipid A (MPL) just before the labelling, in order to initiate the maturation process (subgroup MPL+). Only one MPL+ patient of group A exhibited faint focal uptake in the inguinal region on the late images. Group B presented a more complex pattern of radioactivity distribution (early bladder activity without brain uptake) indicating that 99mTc-HMPAO is not a suitable radiopharmaceutical for labelling of loaded DCs. The activity cleared from DCs as a labelled molecule different from the lipophilic 99mTc-HMPAO. Only one of the six patients had nodular inguinal uptake on the intradermally injected side (DCs not incubated with MPL). In conclusion, the present study did not demonstrate migration of loaded labelled DCs from intradermal or subcutaneous sites of injection to regional lymph nodes. This provides an indication that a large proportion of antigen-loaded DCs, as used in current human trials for cancer therapy, may not reach regional lymph nodes.


Subject(s)
Actins/metabolism , Cell Movement/drug effects , Dendritic Cells/cytology , Dendritic Cells/metabolism , Lymph Nodes/metabolism , Organometallic Compounds/pharmacology , Oxyquinoline/analogs & derivatives , Oxyquinoline/pharmacology , Technetium Tc 99m Exametazime/pharmacology , Antigens/metabolism , Cell Survival/drug effects , Cells, Cultured , Dendritic Cells/diagnostic imaging , Humans , Isotope Labeling/methods , Lymph Nodes/cytology , Lymph Nodes/diagnostic imaging , Organometallic Compounds/pharmacokinetics , Oxyquinoline/pharmacokinetics , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology , Technetium Tc 99m Exametazime/pharmacokinetics
15.
Cardiovasc Surg ; 8(4): 265-73, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10840203

ABSTRACT

Following implantation different cell types interact with synthetic vascular prostheses resulting in a complex immuno-inflammatory response. Dendritic cells are responsible for activating the primary T-lymphocyte immune response in various pathological conditions by their role as antigen-presenters. This study aimed at examining whether dendritic cells accumulate within small diameter expanded polytetrafluoroethylene (ePTFE, Goretex(R)) grafts explanted from humans. Segments of expanded polytetrafluoroethylene were explanted from 11 patients (6 male, 5 female), aged between 60 and 83 years (mean 70.7 years) at secondary or revision bypass operation. The graft implant duration varied from 4 months to 12 years (mean 40.5 months). Dendritic cells were identified immunohistochemically using S-100 antibody, as well as by electron microscopy. Immunohistochemical examination showed that all 11 explanted synthetic grafts contained S-100(+) cells colonising both the synthetic matrix itself, and the adjacent perigraft tissue. Electron microscopic analysis confirmed the presence of cells with a characteristic dendritic cell morphology within the grafts. Dendritic cells which accumulated within synthetic grafts were found to co-localise with T-lymphocytes. Based on these observations, we speculate that dendritic cells may be involved in the immuno-inflammatory responses following the implantation of synthetic vascular prostheses through their interaction with T-lymphocytes.


Subject(s)
Blood Vessel Prosthesis , Dendritic Cells/metabolism , Polytetrafluoroethylene/therapeutic use , Aged , Aged, 80 and over , Dendritic Cells/diagnostic imaging , Female , Humans , Immunohistochemistry , Male , Middle Aged , S100 Proteins/metabolism , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL
...