Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Methods Mol Med ; 29: 123-32, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-21374315

RESUMO

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.

2.
Methods ; 19(1): 187-90, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10525455

RESUMO

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.


Assuntos
Antígenos de Neoplasias/genética , Vacinas Anticâncer/farmacologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Vacinas de DNA/farmacologia , Animais , Vacinas Anticâncer/genética , Expressão Gênica , Vetores Genéticos , Humanos , Técnicas In Vitro , Camundongos , Plasmídeos/genética , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/genética
3.
Br J Cancer ; 79(7-8): 1067-73, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10098738

RESUMO

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.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/análogos & derivados , Imunoterapia Adotiva/métodos , Imunotoxinas/administração & dosagem , Interleucina-2 , Células Matadoras Ativadas por Linfocina , Neoplasias Hepáticas/terapia , Linfoma Difuso de Grandes Células B/terapia , Adolescente , Animais , Antibióticos Antineoplásicos/farmacocinética , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Humanos , Imunotoxinas/farmacocinética , Neoplasias Hepáticas/secundário , Camundongos , Células Tumorais Cultivadas
4.
J Virol ; 73(3): 2280-7, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9971811

RESUMO

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.


Assuntos
Leucemia Experimental/imunologia , Vírus da Leucemia Murina de Moloney/imunologia , Vírus do Sarcoma Murino de Moloney/imunologia , Infecções por Retroviridae/imunologia , Sarcoma Experimental/imunologia , Infecções Tumorais por Vírus/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Animais , Feminino , Produtos do Gene env/imunologia , Produtos do Gene gag/imunologia , Imunização , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas
5.
Br J Cancer ; 77(4): 656-62, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9484826

RESUMO

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.


Assuntos
Hemangiossarcoma/imunologia , Hemangiossarcoma/terapia , Interleucina-12/uso terapêutico , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/terapia , Linfócitos T Citotóxicos/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Hemangiossarcoma/mortalidade , Imunidade Celular , Interferon gama/antagonistas & inibidores , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias Cutâneas/mortalidade , Células Tumorais Cultivadas
7.
Hum Gene Ther ; 8(12): 1451-8, 1997 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-9287145

RESUMO

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.


Assuntos
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Imunização/métodos , Neoplasias/prevenção & controle , Linfócitos T Citotóxicos/imunologia , Animais , Transplante de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos DBA , Neoplasias/genética , Neoplasias/imunologia , Plasmídeos/genética , Plasmídeos/farmacologia , Células Tumorais Cultivadas
8.
Biochem Biophys Res Commun ; 226(3): 769-76, 1996 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-8831688

RESUMO

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.


Assuntos
Trifosfato de Adenosina/farmacologia , Apoptose/fisiologia , Antígenos Comuns de Leucócito/fisiologia , Proteínas Tirosina Fosfatases/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Apoptose/efeitos dos fármacos , Northern Blotting , Morte Celular , Citotoxicidade Imunológica , Citometria de Fluxo , Cinética , Leucemia L1210/enzimologia , Leucemia L1210/imunologia , Antígenos Comuns de Leucócito/biossíntese , Camundongos , Proteínas Tirosina Fosfatases/biossíntese , RNA Mensageiro/biossíntese , Transcrição Gênica , Células Tumorais Cultivadas
9.
Cell Immunol ; 171(1): 102-10, 1996 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-8660844

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , Proteínas de Membrana/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Apoptose/imunologia , Cálcio/imunologia , Cálcio/fisiologia , Linhagem Celular , Testes Imunológicos de Citotoxicidade , Espaço Extracelular/imunologia , Espaço Extracelular/fisiologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/toxicidade
10.
Int J Cancer ; 65(6): 847-51, 1996 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-8631602

RESUMO

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.


Assuntos
Anticorpos Monoclonais/farmacologia , Selectina L/imunologia , Linfonodos/imunologia , Sarcoma Experimental/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Anticorpos Antineoplásicos/imunologia , Divisão Celular/efeitos dos fármacos , Transformação Celular Viral , Linfonodos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Vírus do Sarcoma Murino de Moloney , Ratos , Infecções por Retroviridae/virologia , Sarcoma Experimental/patologia , Sarcoma Experimental/virologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Infecções Tumorais por Vírus/virologia
11.
Biochem Biophys Res Commun ; 218(1): 344-51, 1996 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-8573158

RESUMO

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.


Assuntos
Trifosfato de Adenosina/farmacologia , Apoptose/fisiologia , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular , Dano ao DNA , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Genisteína , Homeostase , Isoflavonas/farmacologia , Leucemia L1210 , Sarcoma de Mastócitos , Camundongos , Fosfotirosina/análise , Proteínas Tirosina Quinases/antagonistas & inibidores , Células Tumorais Cultivadas , Vanadatos/farmacologia
12.
Int J Cancer ; 61(3): 355-62, 1995 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-7729948

RESUMO

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.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Molécula 1 de Adesão Intercelular/imunologia , Antígeno-1 Associado à Função Linfocitária/imunologia , Vírus do Sarcoma Murino de Moloney , Sarcoma Experimental/terapia , Linfócitos T Citotóxicos/imunologia , Animais , Células Cultivadas , Citometria de Fluxo , Expressão Gênica , Molécula 1 de Adesão Intercelular/biossíntese , Contagem de Leucócitos , Antígeno-1 Associado à Função Linfocitária/biossíntese , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/prevenção & controle , Infecções por Retroviridae/terapia , Sarcoma Experimental/imunologia , Sarcoma Experimental/prevenção & controle , Baço/imunologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/prevenção & controle , Infecções Tumorais por Vírus/terapia
13.
Cell Immunol ; 159(2): 294-305, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7994761

RESUMO

Several lines of evidence point to a central role for protein tyrosine kinases (PTKs) in the signal transduction cascade initiated by T-cell receptor (TCR) engagement. In cytotoxic T lymphocytes (CTL), TCR crosslinking leads to activation of the lytic process which includes conjugate formation, lethal hit delivery, and events leading to target cell death. We studied the role of PTKs in antigen-specific cytotoxicity exerted by both in vivo activated and in vitro maintained CTL. We found that the PTK inhibitors herbimycin A and genistein blocked T-cell-mediated lysis in a dose-dependent manner. Lack of cytotoxic function was not due to abrogation of conjugate formation, but was associated with inhibition of both granule exocytosis and phosphatidylinositides turnover, thus indicating that PTK activity is an obligatory event for the activation of antigen-specific CTL effector function.


Assuntos
Citotoxicidade Imunológica/imunologia , Proteínas Tirosina Quinases/fisiologia , Linfócitos T Citotóxicos/imunologia , Animais , Benzoquinonas , Morte Celular/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , DNA/metabolismo , Esterases/metabolismo , Citometria de Fluxo , Genisteína , Fosfatos de Inositol/biossíntese , Isoflavonas/farmacologia , Lactamas Macrocíclicas , Camundongos , Camundongos Endogâmicos DBA , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinonas/farmacologia , Rifabutina/análogos & derivados , Linfócitos T Citotóxicos/efeitos dos fármacos , Células Tumorais Cultivadas
14.
Cell Immunol ; 156(2): 458-67, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8025958

RESUMO

It has been proposed that extracellular ATP (ATPo) may function as a cytotoxic molecule in Ca(2+)-independent cell-mediated lysis by activating plasma membrane P2 purinergic receptors. In the present study the involvement of the P2z purinergic receptor in ATPo as well as cell-mediated lysis was investigated by using the J774 mouse macrophage cell line, which expresses this receptor, and a panel of J774-derived mutant cell clones selected for the lack of P2z receptor activity. We confirmed that the P2z receptor in J774 is associated with ATPo-induced colloido-osmotic lysis but not with apoptosis. Furthermore, we observed that the lack or the inhibition of the P2z purinergic receptor does not affect lytic activity mediated by different types of cytotoxic cell populations. These results on the whole indicate that the P2z receptor is involved in cell membrane damage induced by ATPo but not in cell-mediated cytotoxicity.


Assuntos
Trifosfato de Adenosina/imunologia , Citotoxicidade Imunológica/fisiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Linhagem Celular , DNA/metabolismo , Resistência a Medicamentos , Espaço Extracelular/imunologia , Espaço Extracelular/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Receptores Purinérgicos P2/efeitos dos fármacos , Receptores Purinérgicos P2/imunologia , Receptores Purinérgicos P2/metabolismo , Linfócitos T Citotóxicos/imunologia
15.
Cancer Res ; 54(4): 1016-20, 1994 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-8313356

RESUMO

We studied the potential use of lymphokine-activated killer (LAK) cells loaded with 4'-deoxy-4'-iododoxorubicin (IDX) in adoptive immunotherapy experiments. Because LAK cells preferentially locate in the lung, we evaluated the therapeutic effect of IDX-loaded LAK cells in mice bearing lung metastases induced by B16F1 tumor cell injection. In vitro studies showed that LAK cells rapidly incorporated IDX, with maximum uptake at 15 min, followed by a plateau; drug efflux was initially rapid and then continued at a much slower rate. Evaluation of LAK cell cytotoxic activity against relevant target cells showed a 30% decrease after IDX treatment that progressed with time over the next 6 h. P388 tumor cell growth was inhibited by coculture with IDX-loaded LAK cells, thus demonstrating that the released IDX maintained its pharmacological activity. Finally, high performance liquid chromatography analysis of tissue IDX concentration revealed a considerably higher and long-lasting concentration in the lungs of mice receiving IDX-loaded LAK cells, compared to mice given injections of a comparable amount of free drug. Moreover, adoptive transfer of IDX-loaded LAK cells into tumor-bearing mice caused a significant reduction in the number of lung metastases versus control mice given injections of even higher doses of free drug.


Assuntos
Doxorrubicina/análogos & derivados , Imunoterapia Adotiva , Células Matadoras Ativadas por Linfocina/imunologia , Neoplasias Pulmonares/terapia , Animais , Citotoxicidade Imunológica , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapêutico , Pulmão/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA
16.
Cell Immunol ; 152(1): 110-9, 1993 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8242754

RESUMO

Extracellular ATP (ATPo) was recently considered a possible mediator of cell-mediated cytotoxicity since it is secreted by effector cells following appropriate stimulus and causes lysis as well as DNA degradation of susceptible target cells. This hypothesis however is not readily reconciled with the finding that ATPo-resistant cells are fully susceptible to intact effector cells, which instead suggests that a necessary step in cell-mediated cytotoxicity is the interaction between different molecules released by a cytotoxic cell. By combining ATPo with TNF or lymphotoxin (LT), cytokines which induce late DNA damage, we observed a synergistic effect on target cell death. Under these conditions, the phenomenon of DNA degradation also appeared early, with a kinetics reminiscent of that observed during target cell incubation with intact effector cells. Target cells which are resistant to one of the two molecules exhibited an enhanced rate of cell death when exposed to their association. Target cell lysis and DNA degradation were also Ca(2+)-independent events as they took place following external Ca2+ chelation by EGTA addition and under experimental conditions in which little or no Ca2+ was released from target cell intracellular stores. These findings suggest that ATPo might represent a further mediator which is responsible for the alternative Ca(2+)-independent cytolytic pathway in association with other molecules released by effector cytotoxic lymphocytes.


Assuntos
Trifosfato de Adenosina/farmacologia , DNA/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Cálcio/metabolismo , Morte Celular , Sinergismo Farmacológico , Linfotoxina-alfa/farmacologia , Camundongos , Camundongos Endogâmicos DBA , Timidina/metabolismo , Trítio , Células Tumorais Cultivadas/efeitos dos fármacos , Uridina/metabolismo
19.
Int J Cancer Suppl ; 7: 24-7, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1385341

RESUMO

We have investigated the in vivo role of 2 different adhesion molecules, LFA-1 and LECAM-1, in the immune reaction to Moloney-murine-sarcoma-virus(M-MSV)-induced tumors, which undergo a peculiar spontaneous regression due to generation of a strong virus-specific cytotoxic-T-lymphocyte(CTL) response. Repeated administration of anti-LFA-1 monoclonal antibody (FD441.8 MAb), i.p. or at the site of virus inoculation, enhanced tumor growth and delayed regression, while i.p. administration of anti-LECAM-1 MEL-14 MAb gave rise to tumors that grew progressively and caused host death. Evaluation of the immunological response in MAb-treated mice showed reduced generation of virus-specific CTL precursors (p) in the spleen of animals given FD441.8 MAb i.p.; CTLp frequency in locally treated mice overlapped with that of control mice injected with virus only. FD441.8 MAb treatment did not interfere with CTL homing in the tumor, since the frequency of M-MSV-specific CTLps in sarcomas was similar in treated and control mice. Cytofluorimetric analysis indicated that the majority of tumor-infiltrating lymphocytes (TIL) from MAb-treated mice were covered by anti-LFA-1 MAb, and lacked cytotoxic activity when assayed against target cells bearing relevant tumor antigens. Instead, in mice injected i.p. with MEL-14 MAb, a very low frequency of CTLps was detected in lymph nodes draining the tumor area, and within the tumor. Our results indicate that enhanced tumor growth, depending on the MAb used, is the resultant of an inhibitory effect on different T-lymphocyte functions. Tumor progression in anti-LFA-1 MAb-injected mice is explained mostly by blockage of CTL lytic activity at the tumor site; in mice receiving i.p. MEL-14 MAb treatment, by the failure of naive T lymphocytes to enter peripheral lymph nodes and subsequently by the lack of generation of tumor-specific CTLs.


Assuntos
Moléculas de Adesão Celular/imunologia , Adesão Celular/imunologia , Antígeno-1 Associado à Função Linfocitária/imunologia , Vírus do Sarcoma Murino de Moloney/imunologia , Regressão Neoplásica Espontânea/imunologia , Sarcoma Experimental/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Anticorpos Monoclonais , Citotoxicidade Imunológica , Selectina L , Teste de Cultura Mista de Linfócitos , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Sarcoma Experimental/microbiologia , Células-Tronco/imunologia
20.
Cancer Immunol Immunother ; 35(1): 27-32, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1611620

RESUMO

Adoptive transfer of tumour-specific T lymphocytes loaded with ricin into tumour-bearing mice exerts a transient therapeutic effect against locally induced tumours [Cerundolo et al. (1987) Br J Cancer 55: 413]. As transferred cells preferentially locate in the lung, we studied the therapeutic effect of ricin-loaded, lymphokine-activated killer (LAK) cells on lung metastases induced by M4 or B16-F1 (F1) tumour cell injection. In vitro studies demonstrated that ricin-treated LAK cells were about 100-fold more efficient than untreated LAK cells in inhibiting growth of the ricin-sensitive M4 tumour cell line. This effect was most likely due to the released ricin, as treated and untreated LAK cells inhibited the relatively toxin-resistant F1 cell line to the same extent. Ricin treatment did not alter the tissue distribution of intravenously (i.v.) injected LAK cells, which selectively localized in the lung early after inoculation, whether or not metastases were present. Adoptive transfer experiments showed that ricin-treated LAK cells were significantly more efficient than untreated LAK cells in inhibiting M4- but not F1-induced lung metastases. These results indicate that LAK cells are able to deliver a therapeutic concentration of antineoplastic compounds directly to the lung.


Assuntos
Células Matadoras Ativadas por Linfocina/imunologia , Neoplasias Pulmonares/terapia , Neoplasias Experimentais/terapia , Ricina/administração & dosagem , Animais , Imunização Passiva , Imunoterapia , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos , Transplante de Neoplasias , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA