Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Acta Biomater ; 76: 333-343, 2018 08.
Article in English | MEDLINE | ID: mdl-29966758

ABSTRACT

Mesoporous bioactive glasses (MBGs) in the system SiO2-CaO-P2O5-Ga2O3 have been synthesized by the evaporation induced self-assembly method and subsequent impregnation with Ga cations. Two different compositions have been prepared and the local environment of Ga(III) has been characterized using 29Si, 71Ga and 31P NMR analysis, demonstrating that Ga(III) is efficiently incorporated as both, network former (GaO4 units) and network modifier (GaO6 units). In vitro bioactivity tests evidenced that Ga-containing MBGs retain their capability for nucleation and growth of an apatite-like layer in contact with a simulated body fluid with ion concentrations nearly equal to those of human blood plasma. Finally, in vitro cell culture tests evidenced that Ga incorporation results in a selective effect on osteoblasts and osteoclasts. Indeed, the presence of this element enhances the early differentiation towards osteoblast phenotype while disturbing osteoclastogenesis. Considering these results, Ga-doped MBGs might be proposed as bone substitutes, especially in osteoporosis scenarios. STATEMENT OF SIGNIFCANCE: Osteoporosis is the most prevalent bone disease affecting millions of patients every year. However, there is a lack of bone grafts specifically designed for the treatment of bone defects occurred because of osteoporotic fractures. The consequence is that osteoporotic bone defects are commonly treated with the same biomaterials intended for high quality bone tissue. In this work we have prepared mesoporous bioactive glasses doped with gallium, demonstrating osteoinductive capability by promoting the differentiation of pre-osteoblast toward osteoblasts and partial inhibition of osteoclastogenesis. Through a deep study of the local environment of gallium within the mesoporous matrix, this work shows that gallium release is not required to produce this effect on osteoblasts and osteoclasts. In this sense, the presence of this element at the surface of the mesoporous bioactive glasses would be enough to locally promote bone formation while reducing bone resorption.


Subject(s)
Bone Substitutes , Cell Differentiation/drug effects , Gallium , Glass/chemistry , Osteoblasts/metabolism , Osteoclasts/metabolism , Animals , Bone Substitutes/chemistry , Bone Substitutes/pharmacology , Cell Line , Gallium/chemistry , Gallium/pharmacology , Humans , Mice , Osteoblasts/cytology , Osteoclasts/cytology
2.
Oncogene ; 37(2): 241-254, 2018 01 11.
Article in English | MEDLINE | ID: mdl-28925399

ABSTRACT

Glioblastomas (GBM) are lethal primitive brain tumours characterized by a strong intra-tumour heterogeneity. We observed in GBM tissues the coexistence of functionally divergent micro-territories either enriched in more differentiated and non-mitotic cells or in mitotic undifferentiated OLIG2 positive cells while sharing similar genomic abnormalities. Understanding the formation of such functionally divergent micro-territories in glioblastomas (GBM) is essential to comprehend GBM biogenesis, plasticity and to develop therapies. Here we report an unexpected anti-proliferative role of beta-catenin in non-mitotic differentiated GBM cells. By cell type specific stimulation of miR-302, which directly represses cyclin D1 and stemness features, beta-catenin is capable to change its known proliferative function. Nuclear beta-catenin accumulation in non-mitotic cells is due to a feed forward mechanism between DOCK4 and beta-catenin, allowed by increased GSK3-beta activity. DOCK4 over expression suppresses selfrenewal and tumorigenicity of GBM stem-like cells. Accordingly in the frame of GBM median of survival, increased level of DOCK4 predicts improved patient survival.


Subject(s)
GTPase-Activating Proteins/metabolism , Glioblastoma/pathology , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , beta Catenin/metabolism , Adult , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain/pathology , Cell Nucleus/metabolism , Cell Proliferation , Feedback, Physiological , GTPase-Activating Proteins/genetics , Glioblastoma/genetics , Glioblastoma/mortality , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Mice , Mice, Inbred NOD , MicroRNAs/genetics , Mitosis , Neoplastic Stem Cells/cytology , Oligodendrocyte Transcription Factor 2/metabolism , Primary Cell Culture , RNA, Small Interfering/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult , beta Catenin/genetics
3.
Leukemia ; 29(10): 1993-2002, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25925206

ABSTRACT

The BMI1 polycomb protein regulates self-renewal, proliferation and survival of cancer-initiating cells essentially through epigenetic repression of the CDKN2A tumor suppressor locus. We demonstrate here for the first time that BMI1 also prevents autophagy in chronic myeloid leukemia (CML) cell lines, to support their proliferation and clonogenic activity. Using chromatin immunoprecipitation, we identified CCNG2/cyclin G2 (CCNG2) as a direct BMI1 target. BMI1 downregulation in CD34+ CML cells by PTC-209 pharmacological treatment or shBMI1 transduction triggered CCNG2 expression and decreased clonogenic activity. Also, ectopic expression of CCNG2 in CD34+ CML cells strongly decreased their clonogenicity. CCNG2 was shown to act by disrupting the phosphatase 2A complex, which activates a PKCζ-AMPK-JNK-ERK pathway that engages autophagy. We observed that BMI1 and CCNG2 levels evolved inversely during the progression of CML towards an acute deadly phase, and therefore hypothesized that BMI1 could support acute transformation of CML through the silencing of a CCNG2-mediated tumor-suppressive autophagy response.


Subject(s)
Autophagy , Cell Proliferation , Cyclin G2/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Polycomb Repressive Complex 1/metabolism , Apoptosis , Blotting, Western , Chromatin Immunoprecipitation , Cyclin G2/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , RNA, Small Interfering/genetics , Signal Transduction , Tumor Cells, Cultured
6.
Br J Cancer ; 98(2): 335-44, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18182997

ABSTRACT

NF-kappaB interferes with the effect of most anti-cancer drugs through induction of anti-apoptotic genes. Targeting NF-kappaB is therefore expected to potentiate conventional treatments in adjuvant strategies. Here we used a pharmacological inhibitor of the IKK2 kinase (AS602868) to block NF-kappaB activation. In human colon cancer cells, inhibition of NF-kappaB using 10 microM AS602868 induced a 30-50% growth inhibitory effect and strongly enhanced the action of SN-38, the topoisomerase I inhibitor and CPT-11 active metabolite. AS602868 also potentiated the cytotoxic effect of two other antineoplasic drugs: 5-fluorouracil and etoposide. In xenografts experiments, inhibition of NF-kappaB potentiated the antitumoural effect of CPT-11 in a dose-dependent manner. Eighty-five and 75% decreases in tumour size were observed when mice were treated with, respectively, 20 or 5 mg kg(-1) AS602868 associated with 30 mg kg(-1) CPT-11 compared to 47% with CPT-11 alone. Ex vivo tumour analyses as well as in vitro studies showed that AS602868 impaired CPT-11-induced NF-kappaB activation, and enhanced tumour cell cycle arrest and apoptosis. AS602868 also enhanced the apoptotic potential of TNFalpha on HT-29 cells. This study is the first demonstration that a pharmacological inhibitor of the IKK2 kinase can potentiate the therapeutic efficiency of antineoplasic drugs on solid tumours.


Subject(s)
Camptothecin/analogs & derivatives , Colonic Neoplasms/drug therapy , Drug Delivery Systems , NF-kappa B/antagonists & inhibitors , Pyrimidines/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Camptothecin/administration & dosage , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Female , HT29 Cells , Humans , I-kappa B Kinase/antagonists & inhibitors , Irinotecan , Mice , Mice, Nude , Protein Kinase Inhibitors/administration & dosage , Topoisomerase I Inhibitors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Article in English | MEDLINE | ID: mdl-18003259

ABSTRACT

In this paper, we present the design and implementation of a 3D digital phantom of the neonatal brain. Commonly used digital brain phantoms (e.g. BrainWeb) are based on adults' brains. With the increasing interest in computer aided analysis of neonatal Magnetic Resonance (MR) images, it becomes necessary to create a special digital phantom for neonatal brains. This is because of the pronounced differences not only in size but more important in geometrical proportions of different brain tissues in adults and neonates and the additional need to subdivide the white matter of neonatal brains into two different types. Thus, the here created neonatal brain phantom consists of 6 different tissue types: scalp, skull, gray matter, myelinated and non-myelinated white matter and cerebrospinal fluid. Every voxel has a vector consisting of 6 probabilities of being part of one of these six tissues. The digital brain phantom will be used for simulation of tomographic images of the newborns' head and may serve as well as an evaluation data set for comparison of analysis methods for neonatal MR images, e.g. segmentation/registration algorithms, providing the possibility of controlled degradation of image data.


Subject(s)
Brain/anatomy & histology , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Magnetic Resonance Imaging/methods , Models, Anatomic , Models, Neurological , Phantoms, Imaging , Computer Simulation , Humans , Image Enhancement/methods , Infant, Newborn , Magnetic Resonance Imaging/instrumentation , Reproducibility of Results , Sensitivity and Specificity , Signal Processing, Computer-Assisted
8.
Leukemia ; 21(5): 877-85, 2007 May.
Article in English | MEDLINE | ID: mdl-17330097

ABSTRACT

Acute myeloid leukemia (AML) cells carry molecular defects that promote their leukemic proliferation, resistance to apoptosis and defect in differentiation. Pharmacological targeting of the nuclear factor kappaB (NF-kappaB) pathway has been shown to promote apoptosis of primary AML cells and to sensitize blasts to neoplastic drugs (Frelin, Blood 2005, 105, 804). The Fms-like tyrosine kinase 3 (FLT3), which sustains proliferation of normal hematopoietic progenitors is frequently overexpressed or mutated in AML patients. Using Ba/F3 murine pre-B cells transfected with various mutants of FLT3 (ITD, D835V, D835Y) and the MV4-11 human AML line, we show that normal or oncogenic stimulation of FLT3 led to activation of NF-kappaB. Pharmacological inhibition of either FLT3 with AG1296 or NF-kappaB with the small molecule inhibitor of IkappaB kinase-2 AS602868 reduced viability and triggered cell death. Moreover, AS602868 was also found to interfere directly with FLT3 kinase activation. AS602868 thus appears to target two different kinases that play a crucial role in the pathogenesis of AML, making it particularly attractive as a new therapeutical approach for AML.


Subject(s)
I-kappa B Kinase/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Annexin A5/analysis , Caspase 3/metabolism , Cell Line , Cell Proliferation , Child , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Male , Mice , NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/metabolism , bcl-X Protein/analysis
9.
Am J Pathol ; 154(6): 1867-76, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10362813

ABSTRACT

Transforming growth factor-beta1 (TGF-beta1) has been shown to down-regulate NO synthesis in a variety of normal cells. In the present study, we investigated the influence of TGF-beta1 upon NO production in tumor cells and its consequences for tumor development. During the growth of PROb colon carcinoma cells intraperitoneally injected in syngeneic BDIX rats, intratumoral concentration of TGF-beta1 increases while NO concentration stays very low. Tumor regression induced by intraperitoneal injections of a lipid A is associated with a decrease in TGF-beta1 and an increase in NO intratumoral concentration. In these tumors, PROb tumor cells are the NO- and TGF-beta1-secreting cells. Using PROb cells transfected with an expression vector coding for TGF-beta1 antisense mRNA, we demonstrate in vitro that there is an inverse correlation between the amount of TGF-beta1 secreted and the ability of PROb cells to secrete NO. As the same results were obtained in the presence of an anti-TGF-beta type II receptor neutralizing antibody, and as exogenous TGF-beta1 is without any effect on NO secretion by PROb cells, TGF-beta1 apparently down-regulates NO synthesis in PROb cells by an intracellular mechanism. These results suggest that endogenous TGF-beta1 constitutes a potential target in a search for new antitumoral agents.


Subject(s)
Activin Receptors, Type I , Carcinoma/metabolism , Colonic Neoplasms/metabolism , Intracellular Fluid/metabolism , Nitric Oxide/biosynthesis , Transforming Growth Factor beta/physiology , Animals , Carcinoma/drug therapy , Carcinoma/enzymology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Down-Regulation , Female , Immunotherapy , Lipid A/therapeutic use , Male , Neoplasm Transplantation , Nitric Oxide/pharmacology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Protein Serine-Threonine Kinases/biosynthesis , RNA, Antisense/genetics , RNA, Antisense/pharmacology , Rats , Rats, Inbred Strains , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/biosynthesis , Second Messenger Systems/physiology , Transfection , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured
10.
Cancer Immunol Immunother ; 46(3): 160-6, 1998 May.
Article in English | MEDLINE | ID: mdl-9625539

ABSTRACT

Nitric oxide (NO) has been shown to inhibit the proliferation of lymphocytes. However, in tumour-bearing rats treated with the immunomodulator OM 163, the regressing nodules were heavily infiltrated by T lymphocytes, although they contained high levels of NO. We show here that NO, while inhibiting the proliferation of lymphocytes, increased their life-span, pointing to the ambivalence of this molecule in the course of tumour growth and regression.


Subject(s)
Nitric Oxide/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Adjuvants, Immunologic/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Carcinoma/immunology , Carcinoma/therapy , Cell Survival/drug effects , Escherichia coli , Immunotherapy , Lymphocyte Activation/drug effects , Nitric Oxide/biosynthesis , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/therapy , Rats
11.
J Pharmacol Exp Ther ; 278(2): 926-33, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8768749

ABSTRACT

Although PROb colonic tumor cells are immunosuppressive, the immunomodulator OM 163 induced the disappearance of macroscopic peritoneal nodules in 50% of rats bearing a peritoneal carcinomatosis (p.c.) induced by PROb cells. When the p.c. developed, the number of T lymphocytes was low and the expression of interferon (IFN)-gamma mRNA in tumor nodules decreased very rapidly. The TGF-beta 1 secreted by PROb cells could be responsible for the immunosuppression, because the PROb cell supernatant inhibited IFN-gamma production and T lymphocyte proliferation. When the rats were treated with OM 163, an infiltration of T lymphocytes was observed in tumor nodules, as well as a high expression of the IFN-gamma mRNA. The antitumor efficiency of the immunomodulator OM 163 could be explained in part by a direct effect of OM 163 on T lymphocytes, because in vitro it stimulated the proliferation and the secretion of IFN-gamma of T lymphocytes. OM 163 could also act at the TGF-beta 1 level. Although OM 163 alone or in combination with IFN-gamma did not modify the TGF-beta 1 secretion by PROb cells in vitro, the expression of the TGF-beta 1 mRNA and the TGF-beta 1 protein content was decreased in vivo in treated tumor nodules.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antineoplastic Agents/pharmacology , Down-Regulation/drug effects , Lymphocytes/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Dose-Response Relationship, Drug , Escherichia coli , Gene Expression/drug effects , Immunosuppression Therapy , RNA, Messenger/metabolism , Rats
12.
Biochem Biophys Res Commun ; 209(1): 73-9, 1995 Apr 06.
Article in English | MEDLINE | ID: mdl-7537042

ABSTRACT

The activity of the inducible nitric oxide synthase enzyme (iNOS) is tightly controlled, partly at the transcriptional level. We find NF-kappa B/Rel activation (p50-p50 and p50-p65) in RAW 264.7 macrophages after lipopolysaccharide treatment and binding to both NF-kappa B sites in the mouse iNOS promoter. To delineate the importance of NF-kappa B/Rel in iNOS gene transcription, we used an unusually direct approach to try to improve on the antioxidant-treatment or reporter techniques, namely the depletion of NF-kappa B/Rel activity through the use of a phosphorothioate-modified oligonucleotide containing three copies of the NF-kappa B consensus sequence. The reduction in NF-kappa B/Rel activity (particularly that binding to the downstream of the two sites) was associated with a 50% reduction in NO output and a reduction in the quantity of the iNOS protein expressed. These results point to the probability that physiologically relevant NF-kappa B/Rel activators or repressors other than lipopolysaccharide might crucially affect the macrophage NO response.


Subject(s)
Amino Acid Oxidoreductases/genetics , NF-kappa B/metabolism , Transcription, Genetic , Amino Acid Oxidoreductases/metabolism , Animals , Base Sequence , Binding, Competitive , Gene Expression Regulation, Enzymologic , Mice , Molecular Sequence Data , NF-kappa B p50 Subunit , Nitric Oxide Synthase , Oligodeoxyribonucleotides , Transcription Factor RelA , Transcription Factors/metabolism
13.
Anticancer Res ; 14(3A): 977-9, 1994.
Article in English | MEDLINE | ID: mdl-8074501

ABSTRACT

In a model of colon cancer in rats (peritoneal carcinomatosis), IL-8 was found to have a highly reproducible antitumoural effect. During IL-8-induced tumour regression the infiltration of nodules by CD4+ T lymphocytes was enhanced. However, splenic lymphocytes did not proliferate in response to tumour cells in vitro. IL-8 antitumour effect was associated with a local but not with a systemic activation of T lymphocytes.


Subject(s)
Interleukin-8/therapeutic use , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Neoplasms, Experimental/therapy , T-Lymphocytes/drug effects , Animals , Interleukin-8/pharmacology , Neoplasms, Experimental/immunology , Rats , Rats, Inbred Strains
14.
J Immunol ; 152(10): 5077-83, 1994 May 15.
Article in English | MEDLINE | ID: mdl-7513729

ABSTRACT

The mechanisms of immunosuppression induced by colon cancer in rats were investigated at the systemic and tumor levels. During tumor growth (after i.p. injection of rat colon adenocarcinoma cells in syngeneic BD IX rats), Con A-induced proliferation of splenic mononuclear cells decreased and nitric oxide (NO) production by splenic macrophages increased concomitantly. Incubating splenic mononuclear cells with an inhibitor of NO synthase, NG-monomethyl-L-arginine, restored lymphocyte proliferation. A low level of inducible NO synthase mRNA was detectable in tumors by Northern blotting, with a weak increase during tumor growth. The NO concentration measured in the tumor nodules increased weakly parallel to the tumor growth. Five and six weeks after tumor cell injection, tumor-infiltrating lymphocytes from disaggregated tumors did not proliferate in the presence of Con A. Addition of NG-monomethyl-L-arginine inhibited the production of NO in tumor dissociations and enhanced tumor-infiltrating lymphocyte proliferation. Glyceryl trinitrate (a NO-releasing compound) totally inhibited the lymphocyte proliferation in vitro while it slightly reduced the tumor cell proliferation. T lymphocytes were therefore more sensitive to NO than were tumor cells. Culture medium from tumor cells induced NO production by splenic macrophages, although the factor involved has not yet been identified. Furthermore, tumor cells could also play a part in NO production by tumors because the tumor cells were induced to produce NO by IFN-gamma plus IL-1. These results strongly suggest the participation of NO in the tumor-induced immunosuppression in rats.


Subject(s)
Immune Tolerance , Neoplasms, Experimental/immunology , Nitric Oxide/physiology , Amino Acid Oxidoreductases/biosynthesis , Animals , Cells, Cultured , Female , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Male , Nitric Oxide Synthase , Rats
15.
Immunobiology ; 190(3): 243-54, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8088853

ABSTRACT

The correlation between the activation of macrophages by lipopolysaccharides (LPS) from four different bacterial species and their antitumor effect in a rat model of colon cancer was investigated. The efficacy of LPS from Neisseria meningitidis (Nm), Salmonella minnesota (Sm), Escherichia coli (Ec) and Bordetella pertussis (Bp) was evaluated as the smallest concentration inducing rat peritoneal macrophages (pm psi) to produce tumor necrosis factor (TNF), interleukin-1 (IL-1), IL-6 and nitric oxide (NO). The cytokine production was measured in bioassays and NO production quantitatively with Griess reactant. Nm was the most effective LPS with concentrations of 1 ng/10(6) pm psi for the induction of TNF, IL-1 and IL-6 activities and 0.01 ng/10(6) pm psi for the induction of NO production. The range between efficacy of different LPS was broad from 1 to 10(4)-10(5) for TNF activity, 1 to 10(2)-10(3) for NO production and IL-6 activity and 1 to 10-10(2) for IL-1 activity. In vivo antitumor effect was evaluated on the growth of peritoneal carcinomatosis. Complete tumor regressions were observed, the LPS rating with respect to decreasing efficacy was Nm, Sm, Ec then Bp; Nm, Sm and Ec were very closed while Bp was not effective. These results show the correlation between the antitumor effect in vivo of LPS and their capacity to induce in vitro IL-1 activity, but not between their ability to induce NO production, TNF and IL-6 activities.


Subject(s)
Bacteria/chemistry , Cytokines/biosynthesis , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Neoplasms, Experimental/therapy , Nitric Oxide/biosynthesis , Animals , Carbohydrate Sequence , Cells, Cultured , Female , Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Male , Mice , Mice, Inbred C3H , Molecular Sequence Data , Rats , Rats, Inbred Strains , Tumor Necrosis Factor-alpha/biosynthesis
16.
Cancer Immunol Immunother ; 38(3): 167-70, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8124684

ABSTRACT

The antitumor effect of lipopolysaccharides (LPS) has been observed in several experimental models and is likely to be mediated by macrophages. Stimulation of macrophages with LPS results in the release of several cytokines, including tumor necrosis factor, interleukin-1 and neutrophil-activating peptide-1/interleukin-8 (IL-8), which activates polymorphonuclear leukocytes (PMN) in vitro. Since PMN have an antitumor activity, we tested the in vivo effect of IL-8 on the growth of peritoneal carcinomatoses induced by PROb colon cancer cells in syngeneic rats. IL-8 induced a significant regression of tumors measuring 1-5 mm, and a complete regression was observed in 8 out of 40 rats in four independent experiments. IL-8 was not directly cytotoxic in vitro for tumor cells and was effective in vivo in a narrow range of doses. IL-8 had a significant chemotactic effect for peritoneal PMN in both normal and tumor-bearing rats. PMN taken from the peritoneum of tumor-bearing rats during IL-8 treatment had the same cytotoxic activity against PROb tumor cells as PMN from untreated control rats. Microscopic examinations of tumors during the treatment showed poor infiltrating by PMN. We conclude that the antitumor activity of IL-8 in this model is not mediated by PMN cytotoxicity.


Subject(s)
Adenocarcinoma/drug therapy , Colonic Neoplasms/drug therapy , Interleukin-8/therapeutic use , Neutrophils/immunology , Animals , Antineoplastic Agents , Cytotoxicity, Immunologic , Immunity, Cellular , Rats
17.
Eur J Cancer ; 29A(14): 2003-9, 1993.
Article in English | MEDLINE | ID: mdl-8280496

ABSTRACT

In a model of colon cancer in syngeneic rats, a new immunomodulator, OM 163, induced the complete disappearance of peritoneal carcinomatosis (nodules measuring 1-5 mm) in 41 out of 82 rats. Those results were confirmed in a survival experiment in which 3 out of 10 treated rats died free of tumour 10, 18 and 28 months after the tumour cell injection while all the untreated control rats died of their tumours within 3 months. OM 163 had a systemic effect, since injected intraperitoneally it completely inhibited the growth of lung metastases in 13 out of 20 rats. The antitumour effect of OM 163 was also observed in two rat strains on original tumours. Lymphocyte infiltration was observed in the tumours mainly constituted of CD4+ and CD8+ cells. The treatment had no effect in nude rats, confirming the involvement of T lymphocytes. Furthermore, rats cured by OM 163 were protected against a second challenge of tumour cells and in a Winn's assay, splenocytes from cured rats protected normal rats against tumour cells.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Colonic Neoplasms/therapy , Peritoneal Neoplasms/secondary , T-Lymphocytes/immunology , Animals , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Dose-Response Relationship, Immunologic , Escherichia coli , Female , Lung Neoplasms/secondary , Male , Peritoneal Neoplasms/therapy , Rats , Rats, Inbred F344 , Rats, Nude , Time Factors
18.
Gastroenterology ; 101(3): 726-33, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1860636

ABSTRACT

Colon carcinoma is one of the most frequent causes of cancer death in industrialized countries. The patients generally die of the metastases. In a colon cancer rat model, the authors have shown that lipopolysaccharides from Escherichia coli induced the regression of carcinomatosis and cured 20%-30% of the rats. Some synthetic derivatives of lipid A, which are less toxic than lipopolysaccharides, were injected 14 days after the tumor cells. They induced the complete regression of peritoneal carcinomatosis consisting of numerous nodules measuring 1-5 mm in 20%-30% of rats. Only compounds with three or more hydroxymyristic acid residues were effective. In vivo effects were correlated with the capacity to induce the production of interleukin 1 and tumor necrosis factor but not with the capacity to induce macrophage-mediated cytolysis. It is therefore possible to synthesize weakly toxic derivatives of lipopolysaccharides retaining their antitumoral property in vivo.


Subject(s)
Colonic Neoplasms/therapy , Lipid A/analogs & derivatives , Animals , Colonic Neoplasms/pathology , Female , Interleukin-1/biosynthesis , Lipid A/chemistry , Lipid A/therapeutic use , Macrophages/immunology , Male , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Rats , Tumor Necrosis Factor-alpha/biosynthesis
19.
Int J Cancer ; 48(2): 311-7, 1991 May 10.
Article in English | MEDLINE | ID: mdl-1850388

ABSTRACT

We have studied the effects of 8 cytokines and their combinations on the in vitro growth of 10 human small-cell cancer lines (SCLC). Interferon-alpha and gamma (IFN-alpha and gamma) caused significant but slight growth inhibition over a 7-day incubation period. However, none of the other 6 cytokines, tumor necrosis factor (TNF), lymphotoxin (LT), interleukin-1 beta (IL-1 beta) interleukin-2 (IL-2), transforming growth factor-beta 1 (TGF-beta 1), or granulocyte colony-stimulating factor (G-CSF), modified SCLC cell proliferation. In contrast, all 10 lines were sensitive to lysis by natural killer (NK) and lymphokine-activated killer (LAK) cells. Sensitivity to LAK cells could be increased by pretreatment of SCLC cells with IFN-gamma. As resistance to the cytostatic/cytotoxic activity of some cytokines has been associated with autocrine production of cytokines, we screened the SCLC lines for cytokine mRNAs. Within the limits of detection of the assay we found no expression of TNF, TGF-beta 1, IL-1 beta or IL-6 mRNA in the 10 SCLC lines.


Subject(s)
Carcinoma, Small Cell/therapy , Cytokines/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Lung Neoplasms/therapy , Blotting, Northern , Carcinoma, Small Cell/chemistry , Carcinoma, Small Cell/immunology , Cell Division/drug effects , Cell Division/immunology , Cytokines/genetics , Cytotoxicity Tests, Immunologic , Humans , Interferon-gamma/pharmacology , Lung Neoplasms/chemistry , Lung Neoplasms/immunology , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
20.
Bull Cancer ; 78(3): 249-52, 1991.
Article in English | MEDLINE | ID: mdl-2054523

ABSTRACT

The cloned DHD/K12/TSb line obtained from a chemically-induced rat colon carcinoma, presents tumors which always regress when injected subcutaneously to the syngeneic animal. This study reports that an intraperitoneal injection of the DHD/K12/TSb cells induces a progressive carcinomatosis in a high proportion of the syngeneic rats. This result underlines the effect that the local environment has on tumorigenicity.


Subject(s)
Colonic Neoplasms/pathology , Animals , Cell Line, Transformed/drug effects , Clone Cells , Drug Administration Routes , Female , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Oncogenes , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL