Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Scand J Rheumatol ; 40(2): 139-44, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20919945

ABSTRACT

OBJECTIVES: To compare the distribution of human leucocyte antigen (HLA) class I and II alleles in patients with erosive hand osteoarthritis (EHOA) to that of patients with non-erosive hand OA (non-EHOA) and in healthy Italian Bone Marrow Donors (IBMDs), in order to evaluate possible immunogenetic associations with EHOA. In the EHOA group we also sought possible associations between HLA alleles and disease severity. METHODS: Ninety-four patients with EHOA (82 women, 12 men; mean age 61.4 ± 8.45 years) and 37 with non-EHOA (28 women, nine men; mean age 59.21 ± 9.07 years) were studied. Disease severity was measured by the number of clinically active joints (NCAJ) and by the radiographic score (RS) using the Kallman scale. HLA typing was undertaken for A, B, C, and DRB1 loci; HLA-DRB1* genotyping was determined using polymerase chain reaction (PCR) with sequence-specific primers. Frequencies were compared with those of the healthy IBMDs. RESULTS: The alleles found more frequently in EHOA patients than in non-EHOA patients and healthy controls were: A23, A26, and A29; B38, B44, and HLA DRB1*01 and *07. The RS was more severe in the EHOA compared to the non-EHOA group (63.60 ± 23.14 vs. 34.34 ± 20.24, p < 0.001). Within the EHOA group, HLA-DRB1*07 was associated with a higher RS (67.36 ± 23 vs. 64.5 ± 18.5, p = 0.029). CONCLUSION: In this study of North Italian patients affected with EHOA, the HLA-DRB1*07 allele was found to be associated with both the development and greater severity of the disease.


Subject(s)
Hand Joints/physiopathology , Immunogenetic Phenomena/genetics , Immunogenetic Phenomena/physiology , Osteoarthritis/ethnology , Osteoarthritis/genetics , Aged , Case-Control Studies , Female , Gene Frequency/genetics , HLA-DR Antigens/genetics , HLA-DRB1 Chains , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II/genetics , Humans , Italy , Male , Middle Aged , Osteoarthritis/physiopathology , Severity of Illness Index
2.
Int J Cancer ; 93(1): 85-90, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11391626

ABSTRACT

MAGE, BAGE and GAGE genes encode tumor-associated antigens that are presented by HLA class I molecules and recognized by CD8(+) cytolytic T lymphocytes. These antigens are currently regarded as promising targets for active, specific tumor immunotherapy because MAGE, BAGE and GAGE genes are expressed in many human cancers of different histotype and are silent in normal tissues, with the exception of spermatogonia and placental cells. MAGE, BAGE and GAGE gene expression has been extensively studied in different tumors of adults but is largely unknown in many forms of pediatric solid cancer. Using RT-PCR, we analyzed MAGE-1, MAGE-2, MAGE-3, MAGE-4, MAGE-6, BAGE, GAGE-1,-2 or -8 and GAGE-3,-4,-5,-6 or -7b gene expression in 31 samples of pediatric rhabdomyosarcoma, the most frequent form of malignant soft tissue tumor in children. MAGE genes were expressed in a substantial proportion of patients (MAGE-1, 38%; MAGE-2, 51%; MAGE-3, 35%; MAGE-4, 22%; MAGE-6, 35%), while expression of BAGE (6%); GAGE-1, GAGE-2 and GAGE-8 (9%); and GAGE-3, GAGE-4, GAGE-5, GAGE-6 and GAGE-7B (16%) was less frequent. Overall, 58% of tumors expressed at least 1 gene, and 35% expressed 3 or more genes simultaneously. Our data suggest that a subset of rhabdomyosarcoma patients could be eligible for active, specific immunotherapy directed against MAGE, BAGE and GAGE antigens.


Subject(s)
Antigens, Neoplasm/genetics , Neoplasm Proteins/genetics , Rhabdomyosarcoma/genetics , Humans , Melanoma-Specific Antigens , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/pathology , Tumor Cells, Cultured
3.
Cancer ; 91(10): 1882-8, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11346870

ABSTRACT

BACKGROUND: The MAGE, BAGE, and GAGE gene families code for distinct, tumor specific antigens that are recognized by cytotoxic T lymphocytes in the context of HLA molecules. The purpose of this study was to analyze MAGE, BAGE, and GAGE gene expression in the two major histologic types of esophageal carcinoma, squamous carcinoma (ESCc) and adenocarcinoma (CAc), and to correlate their expression patterns with the principal prognostic parameters and long term survival. METHODS: Gene expression was analyzed in surgical samples from 24 patients with ESCc and 24 patients with CAc by reverse transcriptase-polymerase chain reaction amplification (RT-PCR). None of the patients had received preoperative chemotherapy or radiotherapy, and all were followed until death or for a minimum of 4 years. RESULTS: Sixteen ESCc samples (67%) and 9 CAc samples (37.5%) expressed at least one of the genes under study. The expression of each MAGE gene in the two histologic types was not significantly different, with the exception of MAGE-4, which was expressed more in ESCc samples than in CAc samples. BAGE and GAGE expression was rather low and, in every case, was associated with the expression of at least one MAGE gene. CONCLUSIONS: In the group as a whole, and in both ESCc and CAc subgroups, no significant correlation emerged between the expression of any gene and prognostic parameters, such as pathologic tumor, lymph node, or disease stage. Nevertheless, BAGE or GAGE expression was related significantly to a poor prognosis, whereas the expression of MAGE genes (in the absence of BAGE and GAGE expression) was related significantly to a good prognosis.


Subject(s)
Adenocarcinoma/metabolism , Antigens, Neoplasm/genetics , Carcinoma, Squamous Cell/metabolism , Cardia/metabolism , Esophageal Neoplasms/metabolism , Neoplasm Proteins/genetics , Stomach Neoplasms/metabolism , Adenocarcinoma/pathology , Antigens, Neoplasm/metabolism , Carcinoma, Squamous Cell/pathology , Cardia/pathology , Esophageal Neoplasms/pathology , Female , Gene Expression , Humans , Male , Melanoma-Specific Antigens , Middle Aged , Neoplasm Proteins/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology , Survival Analysis
4.
J Immunother ; 24(6): 431-46, 2001.
Article in English | MEDLINE | ID: mdl-11759067

ABSTRACT

In the late 1970s, several findings suggested that accessory cells distinct from lymphocytes might suppress immune reactivity in tumor-bearing hosts. Studies in animal models and patients later confirmed that cells driven to act as dominant immune suppressors by growing cancers could subvert the immune system. These cells have also been termed natural suppressors, a functional definition connoting their ability to hamper various T- and B-lymphocyte responses without prior activation and independently from antigen and MHC restriction. These properties were attributed to distinct cell populations. The phenotypic discrepancies, together with the lack of antigen specificity, have generated serious restraints to research on tumor-induced suppression. Recent evidence indicates that suppressor cells are closely related to immature myeloid precursors and can be found in several situations that can exert adverse effects on the immunotherapy of cancer. The present review is an attempt to address the nature and properties of immature myeloid suppressors and their relationship to dendritic cells and macrophages, with the aim of clarifying the complex network of tumor-induced, negative regulators of the immune system.


Subject(s)
Immune Tolerance/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Animals , Humans , Receptors, Interleukin-2/immunology , Signal Transduction/immunology
5.
J Immunol ; 165(12): 6723-30, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11120790

ABSTRACT

We described a generalized suppression of CTL anamnestic responses that occurred in mice bearing large tumor nodules or immunized with powerful recombinant viral immunogens. Immune suppression entirely depended on GM-CSF-driven accumulation of CD11b(+)/Gr-1(+) myeloid suppressor cells (MSC) in secondary lymphoid organs. To further investigate the nature and properties of MSC, we immortalized CD11b(+)/Gr-1(+) cells isolated from the spleens of immunosuppressed mice, using a retrovirus encoding the v-myc and v-raf oncogenes. Immortalized cells expressed monocyte/macrophage markers (CD11b, F4/80, CD86, CD11c), but they differed from previously characterized macrophage lines in their capacities to inhibit T lymphocyte activation. Two MSC lines, MSC-1 and MSC-2, were selected based upon their abilities to inhibit Ag-specific proliferative and functional CTL responses. MSC-1 line was constitutively inhibitory, while suppressive functions of MSC-2 line were stimulated by exposure to the cytokine IL-4. Both MSC lines triggered the apoptotic cascade in Ag-activated T lymphocytes by a mechanism requiring cell-cell contact. Some well-known membrane molecules involved in the activation of apoptotic pathways (e.g., TNF-related apoptosis-inducing ligand, Fas ligand, TNF-alpha) were ruled out as candidate effectors for the suppression mechanism. The immortalized myeloid lines represent a novel, useful tool to shed light on the molecules involved in the differentiation of myeloid-related suppressors as well as in the inhibitory pathway they use to control T lymphocyte activation.


Subject(s)
Apoptosis/immunology , Cell Line, Transformed/immunology , Isoantigens/immunology , Lymphocyte Activation/immunology , Macrophages/immunology , Myeloid Cells/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , 3T3 Cells , Animals , Apoptosis Regulatory Proteins , Cell Communication/immunology , Cell Division/immunology , Cell Line, Transformed/metabolism , Cytotoxicity, Immunologic/immunology , Fas Ligand Protein , Female , Immune Tolerance , Immunophenotyping , Ligands , Lymphocyte Culture Test, Mixed , Macrophages/classification , Macrophages/metabolism , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Myeloid Cells/classification , Myeloid Cells/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/physiology , fas Receptor/metabolism
6.
Blood ; 96(12): 3838-46, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11090068

ABSTRACT

Apoptotic death of CD8(+) T cells can be induced by a population of inhibitory myeloid cells that are double positive for the CD11b and Gr-1 markers. These cells are responsible for the immunosuppression observed in pathologies as dissimilar as tumor growth and overwhelming infections, or after immunization with viruses. The appearance of a CD11b(+)/Gr-1(+) population of inhibitory macrophages (iMacs) could be attributed to high levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) in vivo. Deletion of iMacs in vitro or in vivo reversed the depression of CD8(+) T-cell function. We isolated iMacs from the spleens of immunocompromised mice and found that these cells were positive for CD31, ER-MP20 (Ly-6C), and ER-MP58, markers characteristic of granulocyte/monocyte precursors. Importantly, although iMacs retained their inhibitory properties when cultured in vitro in standard medium, suppressive functions could be modulated by cytokine exposure. Whereas culture with the cytokine interleukin 4 (IL-4) increased iMac inhibitory activity, these cells could be differentiated into a nonadherent population of fully mature and highly activated dendritic cells when cultured in the presence of IL-4 and GM-CSF. A common CD31(+)/CD11b(+)/Gr-1(+) progenitor can thus give rise to cells capable of either activating or inhibiting the function of CD8(+) T lymphocytes, depending on the cytokine milieu that prevails during antigen-presenting cell maturation. (Blood. 2000;96:3838-3846)


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Macrophage-1 Antigen/physiology , Macrophages/immunology , Mice, Inbred BALB C , Myeloid Progenitor Cells/immunology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/drug effects , Biomarkers/analysis , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Cytokines/pharmacology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Granulocytes , Immunosuppression Therapy , Interleukin-4/pharmacology , Isoantigens/immunology , Isoantigens/physiology , Lymphocyte Activation/drug effects , Macrophage-1 Antigen/immunology , Macrophages/cytology , Macrophages/drug effects , Mice , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Spleen/cytology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Tumor Cells, Cultured , Vaccinia virus
7.
Cancer Res ; 60(12): 3232-8, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10866316

ABSTRACT

Methoxymorpholinyl doxorubicin (MMDX; PNU 152243) is a promising doxorubicin derivative currently undergoing clinical evaluation. Previous in vitro studies suggested that the compound undergoes hepatic biotransformation by cytochrome P450 (CYP) 3A into a more cytotoxic metabolite(s). The present study examined the role of CYP3A-mediated metabolism in the in vivo antitumor activity and host toxicity of MMDX in the mouse model and investigated the potential for increasing the therapeutic effectiveness of the drug by inducing its hepatic CYP-catalyzed activation. We found that MMDX cytotoxicity for cultured M5076 tumor cells was potentiated 22-fold by preincubating the drug with NADPH-supplemented liver microsomes from untreated C57BL/6 female mice. A greater (50-fold) potentiation of MMDX cytotoxicity was observed after its preincubation with liver microsomes isolated from animals pretreated with the prototypical CYP3A inducer pregnenolone-16alpha-carbonitrile. In contrast, in vivo administration of the selective CYP3A inhibitor troleandomycin (TAO) reduced both potentiation of MMDX cytotoxicity and the rate of CYP3A-catalyzed N-demethylation of erythromycin by isolated liver microsomes (55.5 and 49% reduction, respectively). In vivo antitumor activity experiments revealed that TAO completely suppressed the ability of 90 microg/kg MMDX i.v., a dose close to the LD10, to delay growth of s.c. M5076 tumors in C57BL/6 mice and to prolong survival of DBA/2 mice with disseminated L1210 leukemia. Moreover, TAO administration markedly inhibited the therapeutic efficacy of 90 microg/kg MMDX i.v. in mice bearing experimental M5076 liver metastases; a complete loss of MMDX activity was observed in liver metastases-bearing animals receiving 40 microg/kg MMDX i.v. plus TAO. However, pregnenolone-16alpha-carbonitrile pretreatment failed to enhance MMDX activity in mice bearing either s.c. M5076 tumors or experimental M5076 liver metastases. Additional experiments carried out in healthy C57BL/6 mice showed that TAO markedly inhibited MMDX-induced myelosuppression and protected the animals against lethal doses of MMDX. Taken together, these findings demonstrate that an active metabolite(s) of MMDX synthesized via CYP3A contributes significantly to its in vivo antitumor activity and host toxicity.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/toxicity , Antineoplastic Agents/pharmacology , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/physiology , Doxorubicin/analogs & derivatives , Oxidoreductases, N-Demethylating/physiology , Animals , Anti-Bacterial Agents/pharmacology , Bone Marrow/drug effects , Coculture Techniques , Cytochrome P-450 CYP3A , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Doxorubicin/toxicity , Erythromycin/pharmacology , Female , Leukemia, Experimental/drug therapy , Liver/drug effects , Liver Neoplasms, Experimental/drug therapy , Methylation , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microsomes, Liver/drug effects , NADP/pharmacology , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Pregnenolone Carbonitrile/pharmacology , Protein Synthesis Inhibitors/pharmacology , Time Factors , Troleandomycin/pharmacology , Tumor Cells, Cultured
8.
Cancer Res ; 60(2): 253-8, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10667570

ABSTRACT

"Self" melanocyte differentiation antigens are potential targets for specific melanoma immunotherapy. Vaccination against murine tyrosinase-related protein (TRP)-1/gp75 was shown recently to cause melanoma rejection, which was accompanied by autoimmune skin depigmentation (vitiligo). To further explore the linkage between immunotherapy and autoimmunity, we studied the response to vaccination with a related antigen, TRP-2. i.m. inoculation of plasmid DNA encoding murine trp-2 elicited antigen-specific CTLs that recognized the B16 mouse melanoma and protected the mice from challenge with tumor cells. Furthermore, mice bearing established s.c. B16 melanomas rejected the tumor upon vaccination with a recombinant vaccinia virus encoding trp-2. Depletion experiments showed that CD8+ lymphocytes and natural killer cells were crucial for the antitumor activity of the trp-2-encoding vaccines. Mice that rejected the tumor did not develop generalized vitiligo, indicating that protective immunity can be achieved in the absence of widespread autoimmune aggression.


Subject(s)
Cancer Vaccines/therapeutic use , Interferon Type I/immunology , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Pregnancy Proteins/immunology , Vaccines, Synthetic/therapeutic use , Vitiligo/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/toxicity , Killer Cells, Natural/immunology , Lymphocyte Culture Test, Mixed , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/chemistry , Peptides/immunology , Vaccines, Synthetic/toxicity , Vitiligo/etiology
9.
Methods Mol Med ; 29: 123-32, 2000.
Article in English | MEDLINE | ID: mdl-21374315

ABSTRACT

Many studies have been conducted with the aim to stimulate a therapeutic immune response against tumors. In most cases, efforts have been directed toward the induction of tumor-specific cytotoxic T lymphocytes (CTL), because this T lymphocyte subpopulation is considered to play a major role in the destruction of tumor cells (1). In particular, vaccination protocols have been designed to increase the immunogenicity of intact cancer cells by using adjuvants or engineering tumor cells with cytokine or costimulatory molecule genes. A second line of research has employed immunization with tumor-associated antigens (TAA). These antigens are expressed from derepressed or mutated genes in tumor cells, and are recognized by CTL in the form of peptides associated with MHC class I molecules. Genes encoding TAA have been inserted into recombinant viral vectors, which are then used to infect the host's cells and induce expression of the transgene. Moreover, immunization with purified TAA peptides or with antigen-presenting cells, such as dendritic cells, pulsed with TAA peptides have been proposed.

10.
Methods ; 19(1): 187-90, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10525455

ABSTRACT

Based on experience acquired in the last few years, we describe some technical steps and provide suggestions on how to induce an immune response against tumors expressing the weakly immunogenic antigen P1A by means of a DNA-based vaccination approach. P1A is the product of a normal mouse gene, which shares many characteristics with already identified human tumor-associated antigens, and therefore represents a useful experimental model to evaluate the efficacy of new vaccination strategies potentially applicable to the field of human tumors. Information gained with this model has been applied with success in other experimental settings, and thus we think that the procedure described herein may constitute a valid platform that can be implemented and further refined.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/pharmacology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Vaccines, DNA/pharmacology , Animals , Cancer Vaccines/genetics , Gene Expression , Genetic Vectors , Humans , In Vitro Techniques , Mice , Plasmids/genetics , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/genetics
11.
Br J Cancer ; 79(7-8): 1067-73, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10098738

ABSTRACT

The possibility of using interleukin 2 (IL-2)-activated natural killer cells (A-NK) to carry methoxymorpholinyl doxorubicin (MMDX; PNU 152243) to liver-infiltrating tumours was explored in mice bearing 2-day established M5076 reticulum cell sarcoma hepatic metastases. In vitro, MMDX was 5.5-fold more potent than doxorubicin against M5076 tumour cells. MMDX uptake by A-NK cells correlated linearly with drug concentration in the incubation medium [correlation coefficient (r) = 0.999]; furthermore, as MMDX incorporation was readily reproducible in different experiments, the amount of drug delivered by A-NK cells could be modulated. In vivo experiments showed that intravenous (i.v.) injection of MMDX-loaded A-NK cells exerted a greater therapeutic effect than equivalent or even higher doses of free drug. The increase in lifespan (ILS) following A-NK cell delivery of 53 microg kg(-1) MMDX, a dosage that is ineffective when administered in free form, was similar to that observed in response to 92 microg kg(-1) free drug, a dosage close to the 10% lethal dose (ILS 42% vs. 38% respectively). These results correlated with pharmacokinetic studies showing that MMDX encapsulation in A-NK cells strongly modifies its organ distribution and targets it to tissues in which IL-2 activated lymphocytes are preferentially entrapped after i.v. injection.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/analogs & derivatives , Immunotherapy, Adoptive/methods , Immunotoxins/administration & dosage , Interleukin-2 , Killer Cells, Lymphokine-Activated , Liver Neoplasms/therapy , Lymphoma, Large B-Cell, Diffuse/therapy , Adolescent , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Humans , Immunotoxins/pharmacokinetics , Liver Neoplasms/secondary , Mice , Tumor Cells, Cultured
12.
J Virol ; 73(3): 2280-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-9971811

ABSTRACT

The intramuscular inoculation of Moloney murine sarcoma/leukemia (M-MSV/M-MuLV) retroviral complex gives rise to sarcomas that undergo spontaneous regression due to the induction of a strong immune reaction mediated primarily by cytotoxic T lymphocytes (CTL). We used a DNA-based vaccination approach to dissect the CTL response against the Gag and Env proteins of M-MSV/M-MuLV in C57BL/6 (B6) mice and to evaluate whether plasmid DNA-immunized mice would be protected against a subsequent challenge with syngeneic tumor cells expressing the viral antigens. Intramuscular DNA vaccination induced CTL against both Gag and Env proteins. A detailed analysis of epitopes recognized by CTL generated in mice inoculated with the whole virus and with the Gag-expressing plasmid confirmed the presence of an immunodominant peptide in the leader sequence of Gag protein (Gag85-93, CCLCLTVFL) that is identical to that described in B6 mice immunized with Friend MuLV-induced leukemia cells. Moreover, CTL generated by immunization with the Env-encoding plasmid recognized a subdominant Env peptide (Env189-196, SSWDFITV), originally described in the B6.CH-2(bm13) mutant strain. B6 mice immunized with the Gag-expressing plasmid were fully protected against a lethal tumor challenge with M-MuLV-transformed MBL-2 leukemia cells, while vaccination with the Env-expressing plasmid resulted in rejection of the tumor in 44% of the mice and in increased survival of an additional 17% of the animals. Taken together, these results indicate the existence of a hierarchy in the capacity of different structural viral proteins to induce a protective immune response against retrovirus-induced tumors.


Subject(s)
Leukemia, Experimental/immunology , Moloney murine leukemia virus/immunology , Moloney murine sarcoma virus/immunology , Retroviridae Infections/immunology , Sarcoma, Experimental/immunology , Tumor Virus Infections/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Female , Gene Products, env/immunology , Gene Products, gag/immunology , Immunization , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
13.
Br J Cancer ; 77(4): 656-62, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9484826

ABSTRACT

In syngeneic mice, the H5V polyoma middle-T oncogene-transformed endothelioma cell line induces Kaposi's sarcoma-like cavernous haemangiomas that regress transiently, probably because of an anti-tumour immune response, but eventually grow progressively and kill the host. To evaluate the generation of tumour-specific cytotoxic T lymphocytes (CTLs), spleen cells of tumour-bearing mice were restimulated with irradiated H5V cells in mixed leucocyte-tumour cell cultures. Tumour-specific CTLs were demonstrable only when low numbers of H5V stimulator cells were used (<1 H5V cell per 50 splenocytes). We found that H5V cells secrete immunosuppressive mediators because CTL generation was blocked when H5V cells culture supernatants were added to allogeneic mixed leucocyte cultures. As numerous tumour-derived immunosuppressive mediators may interfere with interleukin 12 (IL-12) production, we tested whether IL-12 treatment of the tumour-bearing mice would augment their immune response and thus suppress tumour growth. Indeed, IL-12 inhibited tumour growth and prevented mortality, but did not increase anti-H5V CTL generation either in vitro or in vivo. Moreover, the anti-tumour activity in IL-12-treated mice was abrogated by anti-interferon (IFN)-gamma monoclonal antibody (MAb) co-administration. These results strongly suggest that the anti-tumour effect of IL-12 is principally mediated by IFN-gamma release that in turn blocks H5V cell proliferation and induces the release of factors that suppress angiogenesis.


Subject(s)
Hemangiosarcoma/immunology , Hemangiosarcoma/therapy , Interleukin-12/therapeutic use , Skin Neoplasms/immunology , Skin Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Monoclonal/pharmacology , Hemangiosarcoma/mortality , Immunity, Cellular , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin Neoplasms/mortality , Tumor Cells, Cultured
15.
Hum Gene Ther ; 8(12): 1451-8, 1997 Aug 10.
Article in English | MEDLINE | ID: mdl-9287145

ABSTRACT

A DNA immunization approach was used to induce an immune response against the tumor-specific antigen P815A in DBA/2 mice. The P1A gene, which encodes the P815A antigen, was modified by the addition of a short sequence coding for a tag epitope recognized by the monoclonal antibody AU1, and cloned into the eukaryotic expression vector pBKCMV, resulting in plasmid pBKCMV-P1A. L1210 cells stably transfected with pBKCMV-P1A expressed P1A mRNA and were lysed by the syngeneic P815A-specific cytotoxic clone CTL-P1:5, thus confirming that the tag-modified P1A protein underwent correct processing and presentation. A single intramuscular injection of 100 microg of pBKCMV-P1A induced the expression of P1A mRNA for at least 4 months. Eighty percent of DBA/2 mice injected three times with 100 microg of pBKCMV-P1A generated cytotoxic T lymphocytes (CTL) that lysed P815 tumor cells, whereas mock-inoculated animals failed to show any cytotoxicity. Moreover, experiments designed to evaluate the protection of pBKCMV-P1A-immunized mice against a lethal challenge with P815 tumor cells showed that 6 of 10 immunized mice rejected the tumor, and 2 mice showed prolonged survival compared to control animals.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Immunization/methods , Neoplasms/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Transplantation , Female , Humans , Mice , Mice, Inbred DBA , Neoplasms/genetics , Neoplasms/immunology , Plasmids/genetics , Plasmids/pharmacology , Tumor Cells, Cultured
16.
Biochem Biophys Res Commun ; 226(3): 769-76, 1996 Sep 24.
Article in English | MEDLINE | ID: mdl-8831688

ABSTRACT

Several lines of evidence implicate protein tyrosine phosphatases (PTP) in the regulation of apoptotic cell death. We have evaluated the role of CD45, the major PTP of hematopoietic cells, in apoptosis induced by extracellular ATP (ATPe) and cytotoxic T lymphocytes (CTL). We observed that two CD45- clones obtained by mutagenesis of the Fas- cell line L1210, exhibit a higher susceptibility to apoptosis induced by ATPe, which was also evident in Ca(2+)-free conditions, when compared to the parental cell line or CD45+ variants. The CD45- cells were also more susceptible to death mediated by an alloreactive CTL clone. When the cytotoxic assay was performed in the presence of EGTA, a Ca2+ chelator, which prevents cytotoxic granule exocytosis and perforin polymerization on target cell membranes, only the CD45- target cells were killed by the CTL clone. These results suggest that a cytotoxic pathway other than the secretory or Fas-dependent pathways was responsible for the enhanced susceptibility of CD45- cells to death, and therefore provide further evidence for the role of ATPe as a possible mediator of Ca(2+)-independent target cell destruction by CTL.


Subject(s)
Adenosine Triphosphate/pharmacology , Apoptosis/physiology , Leukocyte Common Antigens/physiology , Protein Tyrosine Phosphatases/metabolism , T-Lymphocytes, Cytotoxic/immunology , Animals , Apoptosis/drug effects , Blotting, Northern , Cell Death , Cytotoxicity, Immunologic , Flow Cytometry , Kinetics , Leukemia L1210/enzymology , Leukemia L1210/immunology , Leukocyte Common Antigens/biosynthesis , Mice , Protein Tyrosine Phosphatases/biosynthesis , RNA, Messenger/biosynthesis , Transcription, Genetic , Tumor Cells, Cultured
17.
Cell Immunol ; 171(1): 102-10, 1996 Jul 10.
Article in English | MEDLINE | ID: mdl-8660844

ABSTRACT

Tumor necrosis factor alpha, in the secreted as well as membrane-associated (mTNF alpha) form, represents a cytotoxic effector mechanism of activated macrophages; in contrast, direct evidence of the mTNF alpha involvement in cytotoxic T lymphocyte (CTL)-mediated lysis has not yet been obtained. We observed that following activation with anti-CD3 monoclonal antibody (mAb), both cloned CTL and peritoneal exudate lymphocytes rapidly upregulated mTNF alpha; a similar effect was observed in the macrophage cell line J774 after stimulation with lipopolysaccharide endotoxin. Activated effector cells, which were fixed with paraformaldehyde before testing, exerted lytic activity against the TNF-sensitive WEHI 164 tumor cell line, but not against the TNF-resistant P-815 mastocytoma. This effect was completely inhibited in the presence of anti-mouse TNF alpha Ab. Moreover, both mTNF alpha-expressing macrophages and CTL induced nuclear DNA fragmentation in WEHI 164 cells, which was also blocked by anti-TNF alpha Ab and was accompanied by a morphologic degeneration characteristic of the apoptotic form of cell death. These data on the whole indicate a common mode of action for mTNF alpha expressed on different cell populations endowed with cytotoxic capability and also imply a role for this molecule in T-cell-mediated cytotoxicity.


Subject(s)
Apoptosis/drug effects , Cytotoxicity, Immunologic/drug effects , Membrane Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/immunology , Calcium/immunology , Calcium/physiology , Cell Line , Cytotoxicity Tests, Immunologic , Extracellular Space/immunology , Extracellular Space/physiology , Membrane Proteins/metabolism , Membrane Proteins/toxicity , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/toxicity
18.
Int J Cancer ; 65(6): 847-51, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8631602

ABSTRACT

To examine the in vivo contribution of L-selectin in the sensitization of tumor-specific CTL, we investigated the effects of treatment with the anti-L-selectin monoclonal antibody (MAb) MEL-14 on the immune response to Moloney-murine sarcoma virus (M-MSV)-induced tumors, which exhibit spontaneous regression following generation of a strong virus-specific CTL response. Daily systemic administration of MEL-14 for 10 days to M-MSV-injected mice gave rise to larger sarcomas that persisted for a longer time, compared with those arising in control mice injected with virus only. The enhanced tumor growth could not be attributed to cytotoxic activity on leukocytes by MEL-14 since no reduction in the total cell number was detected in peripheral blood and spleen of MAb-treated mice. Evaluation of the immunological response in MAb-treated animals revealed a strong reduction in the generation of virus-specific CTL precursors (CTLp) in tumor-draining peripheral lymph nodes (PLN) 10 and 15 days after M-MSV injection, while in spleen, where lymphocyte localization is independent of L-selectin expression, CTLp generation was only delayed. By day 20, when tumors had begun to regress, the CTLp number showed a marked increase in both spleen and local PLN, where naive recirculating CTL could now enter because L-selectin was no longer down-regulated or blocked by the injected MAb. Our findings indicate that functional inactivation of L-selectin by MEL-14 treatment prevented migration of naive L-selectin+CTL through high endothelial venules (HEV) and their accumulation in PLN draining the tumor area, thereby precluding the initiation of a tumor-specific CTL response that takes place primarily at this site.


Subject(s)
Antibodies, Monoclonal/pharmacology , L-Selectin/immunology , Lymph Nodes/immunology , Sarcoma, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Neoplasm/immunology , Cell Division/drug effects , Cell Transformation, Viral , Lymph Nodes/drug effects , Mice , Mice, Inbred BALB C , Moloney murine sarcoma virus , Rats , Retroviridae Infections/virology , Sarcoma, Experimental/pathology , Sarcoma, Experimental/virology , T-Lymphocytes, Cytotoxic/drug effects , Tumor Virus Infections/virology
19.
Biochem Biophys Res Commun ; 218(1): 344-51, 1996 Jan 05.
Article in English | MEDLINE | ID: mdl-8573158

ABSTRACT

Extracellular ATP (ATPo) induces apoptosis and osmotic lysis in several cell lines. We investigated the role of protein tyrosine kinases (PTKs) and phosphatases (PTPases) in ATPo-induced apoptosis. The PTK inhibitor genistein prevented DNA fragmentation due to ATPo without affecting cell lysis. Comparison of western blot analysis and in vitro kinase assays of anti-phosphotyrosine immunoprecipitates indicated that ATPo activated PTKs whose activity was tightly regulated by PTPases. In fact, an early increase in tyrosine kinase activity was observed after ATPo-treatment and was prevented by specific PTPase inhibitors. In addition, a rapid dephosphorylation of phosphotyrosyl residues on several proteins was detected in ATPo-treated cells. Accordingly, inhibitors of PTPases, but not of serine/threonine phosphatases, were as effective as PTK-inhibitors in blocking ATPo-mediated DNA fragmentation. We describe the early events occurring in ATPo-induced apoptosis and suggest a role for PTPases in cell death.


Subject(s)
Adenosine Triphosphate/pharmacology , Apoptosis/physiology , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line , DNA Damage , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genistein , Homeostasis , Isoflavones/pharmacology , Leukemia L1210 , Mast-Cell Sarcoma , Mice , Phosphotyrosine/analysis , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Cells, Cultured , Vanadates/pharmacology
20.
Int J Cancer ; 61(3): 355-62, 1995 May 04.
Article in English | MEDLINE | ID: mdl-7729948

ABSTRACT

We investigated the effect of combined treatment with anti-LFA-1 and anti-ICAM-1 monoclonal antibodies (MAbs) in the immune reaction to Moloney-murine-sarcoma-virus(M-MSV)-induced tumors, which spontaneously regress due to the generation of a strong virus-specific cytotoxic-T-lymphocyte(CTL) response. Repeated systemic administration of both MAbs to M-MSV-injected mice enhanced tumor growth and delayed regression, while treatment with a single MAb had a similar, though less pronounced, effect. The immune depression achieved could not be attributed to lymphocyte depletion, because no reduction in the total number of leukocytes was detected in the peripheral blood or spleen of these mice. However, anti-LFA-I MAb, alone or in combination with anti-ICAM-I MAb, prevented lymphocyte homing in tumor-draining lymph nodes. Cytofluorimetric analysis disclosed a profound down-modulation of LFA-I and ICAM-I molecule expression on T cells following in vivo MAb treatment. Moreover, in anti-LFA-I MAb-treated mice, the receptor was coated to saturation, while anti-ICAM-I MAb treatment brought about ICAM-I-molecule-coating levels below saturation. Evaluation of M-MSV-specific CTL precursor (p) frequency in lymphoid organs of mice receiving combined MAb treatment showed that CTL generation was greatly reduced 10 days after M-MSV injection, and returned to control levels by day 15. Our findings indicate that systemic administration of MAbs to LFA-I and ICAM-I molecules brings about a strong immune suppressive effect which is mainly due to a block in T-lymphocyte re-circulation, and activation by tumor cells. However, this immune-depressive effect is only temporary, and strictly dependent on continuous MAb administration. Thus, our data suggest that treatment with anti-LFA-I and anti-ICAM-I MAbs combined is unable to induce T-cell tolerance in a highly immunogenic system.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Moloney murine sarcoma virus , Sarcoma, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Cells, Cultured , Flow Cytometry , Gene Expression , Intercellular Adhesion Molecule-1/biosynthesis , Leukocyte Count , Lymphocyte Function-Associated Antigen-1/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred C57BL , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Retroviridae Infections/therapy , Sarcoma, Experimental/immunology , Sarcoma, Experimental/prevention & control , Spleen/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/prevention & control , Tumor Virus Infections/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...