Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J Neuroimmunol ; 117(1-2): 68-77, 2001 Jul 02.
Article in English | MEDLINE | ID: mdl-11431006

ABSTRACT

The purpose of this study was to evaluate mu- and delta-opioid receptors (OR) on human and murine dendritic cells (DC). Expression of mu- and delta-OR mRNA on DC was demonstrated by RT-PCR. The immunocytochemical and Western blot analyses revealed the expression of OR protein in DC. Radioreceptor assay demonstrated the specific saturated temperature-dependent binding of [3H]-labeled opioid ligand on DC and B(max)=2.8+/-0.3 fmol/10(6) cells and K(D)=4.8+/-1.0 nM were calculated by a Scatchard analysis. Finally, OR ligands DADLE and DAGO dose-dependently modulated the capacity of DC to induce T cell proliferation in an MLR assay. Importantly, expression of functional OR on DC was significantly increased upon TNF-alpha-induced DC maturation. Thus, these data suggest a new mechanism of opioid-dependent neuroendocrine immunomodulation.


Subject(s)
Dendritic Cells/chemistry , Receptors, Opioid, delta/analysis , Receptors, Opioid, mu/analysis , Animals , Cells, Cultured , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Messenger/analysis , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/physiology , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/physiology , Tumor Necrosis Factor-alpha/pharmacology
2.
Clin Cancer Res ; 7(3 Suppl): 974s-979s, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300499

ABSTRACT

Melanoma escapes host defenses through a variety of means, including the elimination of immune effector cells within the tumor microenvironment. We have reported recently that murine and human tumors including melanoma induce premature apoptosis of dendritic cells both in vitro and in vivo. In this study, we have demonstrated that overexpression of the Bcl-2 protein family member Bcl-xL rescued murine dendritic cells (DCs) from melanoma-induced death in vitro. Another successful protection approach was tumor necrosis factor (TNF)-alpha-promoted sustained expression of the antiapoptotic protein Bcl-2 within dendritic cells. This effect of TNF-alpha was mediated by inhibition of mitochondrial cytochrome c release. Thus, both Bcl-xL and Bcl-2 enhance survival of dendritic cells within the tumor microenvironment. In addition, mature DCs were more resistant to melanoma-induced apoptosis than immature dendritic cells. This finding suggests a stage-dependent sensitivity of DCs to tumor-induced cell death. We conclude that: (a) mature DCs might be more suitable for the use of cancer vaccination; and (b) Bcl-2 protein family members such as Bcl-xL and Bcl-2 rescue DCs from tumor-induced premature apoptosis.


Subject(s)
Apoptosis , Cytochrome c Group/antagonists & inhibitors , Dendritic Cells/metabolism , Melanoma/pathology , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Blotting, Western , Cytochrome c Group/metabolism , DNA Fragmentation , Male , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Transfection , Tumor Cells, Cultured , bcl-X Protein
3.
Prostate ; 46(1): 68-75, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11170134

ABSTRACT

BACKGROUND: The progression of prostate cancer is accompanied by a marked suppression of the immune system, including the apoptotic death of dendritic cells (DC) responsible for the induction of antitumor immunity. In this study, we evaluated whether prostate cancer might inhibit DC generation and maturation in vitro. METHODS: DC were generated from peripheral blood monocytes in the presence of the human prostate cell line LNCaP or nonmalignant cells, and characterized by light microscopy, FACScan analysis, and ability to stimulate T-cell proliferation. RESULTS: Prostate cancer significantly inhibited the conversion of monocytes into DC, which was assessed by the expression of DC markers CD1a and CD83. These cells were weak stimulators of T-cell proliferation, suggesting that DC generated in the prostate cancer microenvironment are functionally inhibited. CONCLUSIONS: Prostate cancer not only kills mature DC, but also inhibits their generation and maturation, resulting in decreased production of antigen-presenting cells and inhibition of their functional activity.


Subject(s)
Dendritic Cells/immunology , Prostatic Neoplasms/immunology , Antigens, CD , Antigens, CD1/analysis , Dendritic Cells/pathology , Flow Cytometry , Histocytochemistry , Humans , Immunoglobulins , Lymphocyte Culture Test, Mixed , Male , Membrane Glycoproteins , Monocytes/immunology , Monocytes/pathology , Prostatic Neoplasms/pathology , Scintillation Counting , Tritium , Tumor Cells, Cultured , CD83 Antigen
4.
Prostate Cancer Prostatic Dis ; 4(4): 221-227, 2001.
Article in English | MEDLINE | ID: mdl-12497022

ABSTRACT

We have recently shown that human prostate cancer (PCa) cells induced apoptotic death of the most potent antigen-presenting cells, dendritic cells (DC), which are responsible for the induction of specific antitumor immune responses. Here we have evaluated the effect of murine PCa cells RM-1 on the survival of immature and tumor necrosis factor-alpha (TNF-alpha)-stimulated mature DC. PCa cells and DC were co-incubated for 24-48 h and DC apoptosis was assessed by morphologic criteria, Annexin V assay, and TUNEL staining. We have shown that co-incubation of RM-1 cells with DC is accompanied by an increased level of DC apoptosis, which was mediated by decreased expression of anti-apoptotic protein Bcl-2. Stimulation of DC maturation by TNF-alpha resulted in increased resistance of DC to PCa-induced apoptosis. In TNF-alpha treated mature DC, but not in immature DC, the expression of Bcl-2 was not blocked after exposure to RM-1-derived factors. Thus, these data suggest that TNF-alpha-induced maturation of DC increases their resistance to PCa induced apoptosis. This is likely to be due to the stabilizing of the expression of anti-apoptotic protein Bcl-2. The difference in the sensitivity of mature and immature DC to PCa-induced cell death should be considered during the design of DC-based clinical trials for PCa patients.Prostate Cancer and Prostatic Diseases (2001) 4, 221-227.

5.
Forum (Genova) ; 10(3): 204-26, 2000.
Article in English | MEDLINE | ID: mdl-11007930

ABSTRACT

Incidence and mortality of human malignant melanoma has risen rapidly over recent decades. Systemic therapies for metastatic cutaneous melanoma, the most aggressive of all skin cancers, remain disappointing although immunological treatment has been more successful for melanoma than for most other tumours. With the availability of recombinant cytokines, immunotherapy for melanoma has entered a new era and a growing body of evidence suggests the efficacy of these approaches in pre-clinical models. Cytokine gene transfer to tumour cells has been demonstrated to induce tumour rejection in different murine melanoma models suggesting that vaccination with tumour cells genetically engineered to produce cytokines is an attractive strategy to enhance anti-tumour immune responses in patients with melanoma. Taken together, these data may hold significant promise for the development of effective ex vivo and in vivo systemic and gene therapy modalities to counter the highly metastatic nature of human melanoma.


Subject(s)
Immunotherapy , Melanoma, Experimental/therapy , Melanoma/therapy , Skin Neoplasms/therapy , Animals , Cancer Vaccines/administration & dosage , Gene Transfer Techniques , Genetic Therapy , Granulocyte Colony-Stimulating Factor/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Interferons/therapeutic use , Interleukin-1/administration & dosage , Interleukin-1/therapeutic use , Interleukin-12/administration & dosage , Interleukin-12/therapeutic use , Interleukin-18/administration & dosage , Interleukin-18/therapeutic use , Interleukin-2/administration & dosage , Interleukin-2/therapeutic use , Interleukin-4/administration & dosage , Interleukin-4/therapeutic use , Interleukin-6/administration & dosage , Interleukin-6/therapeutic use , Macrophage Colony-Stimulating Factor/therapeutic use , Melanoma/genetics , Melanoma/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Mice, Nude , Neoplasm Metastasis , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Time Factors , Tumor Cells, Cultured/drug effects , Tumor Necrosis Factor-alpha/therapeutic use
6.
Eur J Immunol ; 30(9): 2565-75, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11009090

ABSTRACT

Interleukin-12 (IL-12) and Flt3 ligand (FL) regulate hematopoiesis by stimulating proliferation, differentiation and migration of progenitor and/or stem cells. In this study, we evaluated the in vivo effect of IL-12 alone or in combination with FL on dendritic cell (DC) generation and accumulation in murine spleen, lymph node, thymus, liver, and tumor tissues. Both cytokines induced accumulation of functional DC in lymphoid and non-lymphoid tissues. IL-12 promoted predominantly myeloid dendropoiesis, while FL induced both myeloid and lymphoid dendropoiesis. Combination treatment resulted in a dramatic increase in CD86+, and particularly, NLDC-145+ cells within the liver, which was largely due to cell proliferation. Combination therapy also revealed the ability of FL to protect bone marrow cell populations from IL-12-induced depletion in vivo. In vitro, we found a significant FL-induced up-regulation of IL-12 production by DC at both mRNA and protein levels. Thus, our study suggests that (i) the antitumor activity of IL-12 may, at least in part, be mediated by the stimulation of dendropoiesis and (ii) IL-12 might contribute to the antitumor activity of FL. Furthermore, induction of DC generation in vivo by a combination of IL-12 and FL might become a new approach for immunotherapy of cancer.


Subject(s)
Dendritic Cells/drug effects , Interleukin-12/pharmacology , Membrane Proteins/pharmacology , Animals , Bone Marrow Cells/drug effects , Cells, Cultured , Dendritic Cells/physiology , Interleukin-12/biosynthesis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Organ Specificity , Spleen/cytology , Spleen/drug effects , T-Lymphocytes/physiology
7.
J Immunol ; 165(4): 1956-64, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10925278

ABSTRACT

We have shown that prostate cancer (PCa) causes apoptosis of dendritic cells (DC), which might block the development of specific antitumor immune responses. Analysis of murine prostatic carcinoma tissues revealed the significant decrease in intratumoral DC number during tumor progression. We demonstrated that the cytokine-mediated increase in DC survival was accompanied by an elevated expression of the anti-apoptotic protein Bcl-xL. Next, we evaluated the resistance to tumor-induced apoptosis and the antitumor efficiency of genetically engineered DC overexpressing Bcl-xL. DC were transduced with an adenoviral vector encoding the murine Bcl-xL gene and injected intratumorally. Data analysis revealed that treatment of PCa-bearing mice with Bcl-xL-transduced DC resulted in significant inhibition of tumor growth compared with the administration of nontransduced DC. Thus, our data suggest that the protection of DC from PCa-induced apoptosis might significantly increase the efficacy of DC-based therapies in cancer even in the absence of available tumor-specific Ags.


Subject(s)
Apoptosis/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Proto-Oncogene Proteins c-bcl-2/genetics , Transfection/immunology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Cell Count , Cell Movement/immunology , Cell Survival/immunology , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Disease Progression , Fas Ligand Protein , Gene Expression Regulation/immunology , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Genetic Vectors/therapeutic use , Growth Inhibitors/administration & dosage , Growth Inhibitors/genetics , Growth Inhibitors/immunology , Growth Inhibitors/therapeutic use , Humans , Immunity, Innate , Injections, Intralesional , Interleukin-12/administration & dosage , Interleukin-12/biosynthesis , Interleukin-12/genetics , Interleukin-12/therapeutic use , Ligands , Male , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Neoplasm Transplantation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Proto-Oncogene Proteins c-bcl-2/administration & dosage , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/therapeutic use , Tumor Cells, Cultured , bcl-X Protein , fas Receptor/metabolism , fas Receptor/physiology
8.
Br J Cancer ; 83(4): 506-13, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10945499

ABSTRACT

Prostate cancer is the most common cancer in men in the United States, and second in cancer-induced mortality. It is likely that tumour-induced immunosuppression is one of the reasons for low treatment efficacy in patients with advanced prostate cancer. It has been recently demonstrated that prostate cancer tissue is almost devoid of dendritic cells (DC), the major antigen-presenting cells responsible for the induction of specific antitumour immune responses. In this study, we have tested the hypothesis that prostate cancer induces progressive suppression of the DC system. We found that co-incubation of human DC with three prostate cancer cell lines led to the high levels of premature apoptosis of DC, which were significantly higher than in DC cultures co-incubated with normal prostate cells or blood leucocytes. Stimulation of DC for 24 hours with CD40 ligand (CD154), IL-12 or IL-15 prior to their co-incubation with prostate cancer cells resulted in a significant increase in DC survival in the tumour microenvironment. Furthermore, activation of DC with these cytokines was also accompanied by increased expression of the anti-apoptotic protein Bcl-x(L) in DC, suggesting a possible mechanism involved in DC protection from apoptotic death. In summary, our data demonstrate that prostate cancer induces active elimination of DC in the tumour microenvironment. Stimulation of DC by CD154, IL-12 or IL-15 leads to an increased expression of the anti-apoptotic protein Bcl-x(L) and increased resistance of DC to prostate cancer-induced apoptosis. These results suggest a new mechanism of tumour escape from immune recognition and demonstrate the cytokine-based approaches which might significantly increase the efficacy of DC-based therapies for cancer.


Subject(s)
Apoptosis/physiology , Cell Communication/physiology , Dendritic Cells/pathology , Interleukin-2/physiology , Interleukin-5/physiology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , CD40 Antigens/genetics , CD40 Antigens/physiology , CD40 Ligand , Coculture Techniques , Dendritic Cells/metabolism , Humans , Interleukin-2/biosynthesis , Interleukin-5/biosynthesis , L Cells , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Transfection , Tumor Cells, Cultured , Up-Regulation , bcl-X Protein
9.
Cancer J Sci Am ; 6 Suppl 1: S61-6, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10685662

ABSTRACT

PURPOSE: Recombinant interleukin (IL)-2 administration can mediate regression of solid tumors in patients with melanoma and renal cell carcinoma. A better understanding of the mechanisms of IL-2-mediated antitumor effects has led to the investigation of novel immunotherapeutic approaches. The rationale for these immunotherapeutic approaches and the results of preliminary clinical studies are presented. PATIENTS AND METHODS: The therapeutic potential of dendritic cells and the role of FLT3 ligand, a potent hematopoietic growth factor, was investigated in a variety of preclinical models. In addition, a clinical study with autologous dendritic cells pulsed with synthetic melanoma peptides derived from the MART1/ Melan A, gp100, and tyrosinase proteins was conducted. Twenty-eight human leukocyte antigen (HLA)-A2+ melanoma patients received an average of 106 dendritic cells a week for 4 weeks. RESULTS: In a murine liver metastases model, FLT3 ligand administration alone or in combination with IL-12 or IL-2 had significant antitumor effects and resulted in significant infiltration of the tumor border by lymphocytes and dendritic cells, which was associated with an increased number of apoptotic figures. Administration of melanoma peptide-pulsed dendritic cells to 28 patients with advanced metastatic melanoma produced a complete response in two patients and a partial response in one. Significant infiltration of T cells and dendritic cells into melanoma lesions was observed. CONCLUSION: These studies confirm the feasibility of immunotherapeutic approaches using dendritic cells and FLT3 ligand and demonstrate their potential antitumor activity. These approaches may be effective for patients with metastatic melanoma and other solid tumors and will likely be used to improve the efficacy of IL-2-based immunotherapy.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Genetic Therapy , Interleukin-12/therapeutic use , Interleukin-2/therapeutic use , Membrane Proteins/therapeutic use , Neoplasms/therapy , Fibroblasts/metabolism , Humans , Melanoma/therapy , Neoplasms/immunology
10.
J Invest Dermatol ; 113(6): 1028-32, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10594747

ABSTRACT

An abrogation of the decline in epidermal Langerhans cell numbers above melanoma might significantly improve the efficacy of immunotherapy for melanoma treatment. Systemic Flt3 ligand (FL) administration in mice induced a significant increase in mature dendritic cells (DC) within the skin, preferentially in the dermis, whereas IL-12 promoted a significant increase of immature DC preferentially in the epidermis. Both effects were abrogated in IL-12 knockout mice. Thus, IL-12 could promote FL-induced accumulation of skin DC. The involvement of FL and IL-12 in the regulation of DC accumulation within the skin may contribute, at least in part, to the stimulation of antimelanoma immunity by FL- and IL-12-based immunotherapies. Moreover, FL and IL-12 could be used for selective in vivo generation of DC in either epidermis or dermis for experimental and clinical purposes.


Subject(s)
Dendritic Cells/drug effects , Epidermis/drug effects , Interleukin-12/pharmacology , Membrane Proteins/pharmacology , Skin/drug effects , Animals , Drug Synergism , Langerhans Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
J Immunol ; 163(6): 3260-8, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10477595

ABSTRACT

The cytokine FLT3 ligand (FL) enhances dendritic cell (DC) generation and has therefore been proposed as a means to boost antitumor immunity. Vascular endothelial growth factor (VEGF) is produced by a large percentage of tumors and is required for development of tumor neovasculature. We previously showed that VEGF decreases DC production and function in vivo. In this study, we tested the hypothesis that VEGF regulates FL effects on DC generation. In seven experiments, four groups of mice were treated with PBS, VEGF alone (100 ng/h), FL alone (10 microgram/day), or with the combination of FL and VEGF. VEGF and PBS were administered continuously for 14 days via s.c. pumps. FL was given s.c. daily for 9 days, beginning on day 4. Tissues were collected and the number, phenotype, and function of lymph node, splenic, and thymic DCs were analyzed on day 14. As expected, treatment with FL resulted in a marked increase in the number of lymph node and spleen DCs and a smaller increase in thymic DC. Pretreatment of mice with VEGF inhibited these FL effects in lymph nodes and thymus by about 50%, whereas spleen DC numbers were undiminished by VEGF. VEGF treatment in vivo also inhibited the ability of FL to increase the number of hemopoietic precursor cells and the level of maturity exhibited by DC derived from these hemopoietic precursor cells in vitro. VEGF inhibited FL-inducible activation of transcription factor NF-kappaB. These data suggest that VEGF interferes with the ability of FL to promote dendritic cell differentiation from bone marrow progenitor cells in mice and therefore may decrease the therapeutic efficacy of FL in settings where increased numbers of DCs might provide clinical benefits.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Endothelial Growth Factors/administration & dosage , Lymphokines/administration & dosage , Membrane Proteins/administration & dosage , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Division/drug effects , Cell Division/immunology , Cells, Cultured , Dendritic Cells/drug effects , Endothelial Growth Factors/physiology , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Infusion Pumps , Injections, Subcutaneous , Ligands , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphokines/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Spleen/cytology , Spleen/immunology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
J Leukoc Biol ; 66(2): 336-44, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10449178

ABSTRACT

The induction of apoptosis in T cells is one of several mechanisms by which tumors escape immune recognition. We have investigated whether tumors induce apoptosis in dendritic cells (DC) by co-culture of murine or human DC with different tumor cell lines for 4-48 h. Analysis of DC morphological features, JAM assay, TUNEL, caspase-3-like and transglutaminase activity, Annexin V binding, and DNA fragmentation assays revealed a time- and dose-dependent induction of apoptosis in DC by tumor-derived factors. This finding is both effector and target specific. The mechanism of tumor-induced DC apoptosis involved regulation of Bcl-2 and Bax expression. Double staining of both murine and human tumor tissues confirmed that tumor-associated DC undergo apoptotic death in vivo. DC isolated from tumor tissue showed significantly higher levels of apoptosis as determined by TUNEL assay when compared with DC isolated from spleen. These findings demonstrate that tumors induce apoptosis in DC and suggest a new mechanism of tumor escape from immune recognition. DC protection from apoptosis will lead to improvement of DC-based immunotherapies for cancer and other immune diseases.


Subject(s)
Apoptosis/immunology , Dendritic Cells/immunology , Neoplasms/immunology , Animals , Caspase 3 , Caspases/metabolism , Cells, Cultured , Coculture Techniques , DNA Fragmentation , Electrophoresis, Agar Gel , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/metabolism , Transglutaminases/metabolism , Tritium , Tumor Cells, Cultured
15.
Eur J Immunol ; 29(7): 2148-55, 1999 07.
Article in English | MEDLINE | ID: mdl-10427977

ABSTRACT

We have recently demonstrated that murine and human tumors induce apoptosis of dendritic cells (DC). Here, we evaluated the effect of CD40 ligation on the survival of tumor-associated DC and tumor growth. Retroviral transduction of MC38 colon carcinoma cells with the CD154 gene resulted in inhibition of tumor growth. This effect was abrogated in IL-12 knockout mice. Immunohistochemical analysis revealed an increase in CD11c+ (N418) and CD8+ but not NLDC-145+ cells in CD154-transfected tumors in wild-type mice. This increase was less pronounced in IL-12-deficient mice. In vitro, overexpression of CD154 on tumor cells significantly decreased the level of tumor-induced DC apoptosis. Surprisingly, the CD154-induced protection of DC from tumor-induced apoptosis was IL-12 independent in vitro, suggesting an IL-12-dependent and an IL-12-independent mechanism of CD154-induced anti-tumor immunity. Thus, our data suggest a new strategy to improve immunotherapy of cancer by protecting DC from tumor-induced apoptosis.


Subject(s)
Apoptosis/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Membrane Glycoproteins/physiology , Animals , CD40 Ligand , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Colonic Neoplasms/therapy , Humans , Immunotherapy , Integrin alphaXbeta2/metabolism , Interleukin-12/genetics , Interleukin-12/physiology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Transduction, Genetic
16.
J Environ Monit ; 1(5): 459-64, 1999 Oct.
Article in English | MEDLINE | ID: mdl-11529164

ABSTRACT

Recent studies have demonstrated the utility of ultrasonic extraction (UE), followed by portable anodic stripping voltammetry (ASV), for the on-site determination of lead in environmental and industrial hygiene samples. The aim of this work was to conduct an interlaboratory evaluation of the UE-ASV procedure, with a goal of establishing estimates of method performance based on results from collaborative interlaboratory analysis. In this investigation, performance evaluation materials (PEMs) with characterized lead concentrations were used for interlaboratory testing of the UE-ASV procedure. The UE-ASV protocol examined has been promulgated in the form of two separate national voluntary consensus standards (one for UE and another for electroanalysis, which includes ASV). The PEMs consisted of characterized and homogenized paints, soils, and dusts (the last of which were spiked onto wipes meeting national voluntary consensus standard specifications), and air filter samples (mixed cellulose ester membrane) generated using characterized paints within an aerosol chamber. The lead concentrations within the PEMs were chosen so as to bracket pertinent action levels for lead in the various sample matrices. The interlaboratory evaluation was conducted so as to comply with an applicable national voluntary consensus standard that can be used to estimate the interlaboratory precision of a given analytical test method. Based on the analytical results reported by the participating laboratories, relative standard deviations (RSDs) for repeatability and reproducibility were computed for three different lead contents of the four PEMs. RSDs for repeatability were 0.019-0.100 for paints; 0.030-0.151 for soils; 0.085-0.134 for dust wipes; and 0.095-0.137 for air filters. RSDs for reproducibility were 0.127-0.213 for paints; 0.062-0.162 for soils; 0.085-0.134 for dust wipes; and 0.114-0.220 for air filters. With the exception of one of the air filter samples and one of the paint samples, the precision estimates were within the +/- 20% precision requirement specified in the US Environmental Protection Agency National Lead Laboratory Accreditation Program (NLLAP). The results of this investigation illustrate that the UE-ASV procedure is an effective method for the quantitative measurement of lead in the matrices evaluated in this study.


Subject(s)
Air Pollution, Indoor/analysis , Environmental Monitoring/instrumentation , Lead/analysis , Dust , Electrodes , Environmental Exposure , Environmental Monitoring/methods , Housing , Humans , Paint , Reproducibility of Results , Ultrasonics
17.
Curr Opin Mol Ther ; 1(1): 72-81, 1999 Feb.
Article in English | MEDLINE | ID: mdl-11249687

ABSTRACT

Dendritic cells (DC) are the most potent antigen presenting cells (APC) and the only ones capable of presenting novel antigens to naïve T-cells. Large numbers of DC can be generated in vitro in the presence of appropriate cytokine cocktails using either adherent peripheral blood mononuclear cells (PBMC) or CD34+ precursors. More than 20 preclinical studies have demonstrated the effectiveness of antigen-loaded DC to mediate antitumor immune responses. Three clinical trials have been reported to date that show DC as a promising tool for the immunotherapy of cancer. However, completion and analysis of randomized trials to establish the appropriate antigen(s), adjuvant(s), dose, route and schedule will be crucial. Future DC-based therapies will include genetic modification of DC, the use of CD34+ precursors, direct delivery of DC to tumors, and application of tumor lysates or apoptotic cells as sources of additional, as yet undefined, antigens.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Animals , Antigen Presentation , Antigens, Neoplasm , Cell Differentiation , Clinical Trials as Topic , Dendritic Cells/cytology , Humans , Immunotherapy , In Vitro Techniques , Mice , Neoplasms/immunology , Neoplasms/therapy
19.
J Immunol ; 161(11): 6164-70, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9834102

ABSTRACT

FLT3-ligand (FL) is a recently described cytokine that stimulates the proliferation and differentiation of hematopoietic progenitors both in vivo and in vitro and, when administered to mice, induces an accumulation of dendritic cells (DC) in different lymphoid and nonlymphoid organs and tissues, including the liver. We have studied the antitumor effect of FL administered alone or in combination with IL-12 in a day 3 murine liver metastasis model. FL significantly reduced the number of hepatic metastases (36.00 +/- 11.00 vs 92.00 +/- 10.19 in control group, p < 0.05). Histologic evaluation of the livers revealed that FL induced a significant infiltration of the tumor border by lymphocytes and DC associated with increased number of apoptotic figures. Immunohistochemical analysis demonstrated that FL significantly enhanced the number of DC in the liver parenchyma and within the liver metastases, as well as the number of CD4+ and CD8+ T lymphocytes. These data support the suggestion that DC may be directly involved in the antitumor effect of FL. Interestingly, the antitumor effect of FL was greatly reduced by the NK depletion. Combination of FL and IL-12 resulted in greater antitumor efficacy than these cytokines alone. In summary, we have shown that FL has significant antitumor effect on preexisting murine C3 liver tumors that is mediated by NK cells. We have also demonstrated that the FL/IL-12 combination has an enhanced antitumor activity in the same murine tumor model.


Subject(s)
Antineoplastic Agents/administration & dosage , Killer Cells, Natural/immunology , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Membrane Proteins/administration & dosage , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Count , Cell Movement/immunology , Dendritic Cells/pathology , Growth Inhibitors/administration & dosage , Growth Inhibitors/therapeutic use , Injections, Intraperitoneal , Interleukin-12/therapeutic use , Ligands , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Lymphocyte Count , Lymphocytes/pathology , Macrophages/pathology , Male , Membrane Proteins/therapeutic use , Mice , Mice, Inbred C57BL , Sarcoma, Experimental/immunology , Sarcoma, Experimental/pathology , Sarcoma, Experimental/prevention & control , Sarcoma, Experimental/secondary , Tumor Cells, Cultured
20.
Cytokine Growth Factor Rev ; 9(1): 37-48, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9720755

ABSTRACT

Flt3 ligand (FL) is a recently identified cytokine having a central role in the proliferation, survival and differentiation of early murine and human hematopoietic precursor/stem cells. FL acts synergistically in vitro with a number of other hematopoietic growth factors such as IL-3, IL-6, IL-11, IL-12, KIT Ligand and GM-CSF. Recently, it has been shown the in vivo administration of FL results in a significant alteration of hematopoiesis in murine bone marrow (BM), spleen, peripheral blood, liver and lymph nodes. In addition, treatment with FL resulted in a significant accumulation of functionally active dendritic cells within murine lymphoid tissues. The possible applications of FL in dendritic cell-based immunotherapies are discussed.


Subject(s)
Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Humans , Ligands , Membrane Proteins/genetics , Membrane Proteins/therapeutic use , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , fms-Like Tyrosine Kinase 3
SELECTION OF CITATIONS
SEARCH DETAIL
...