Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
BMC Cancer ; 22(1): 845, 2022 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-35922755

RESUMO

BACKGROUND: Although immune-checkpoint inhibitors (ICI) are overall promissory for cancer treatment, they entail, in some cases, an undesired side-effect called hyperprogressive-cancer disease (HPD) associated with acceleration of tumor growth and shortened survival. METHODS: To understand the mechanisms of HPD we assayed the ICI therapy on two murine tumors widely different regarding immunogenicity and, subsequently, on models of local recurrences and metastases of these tumors. To potentiate the immune response (IR), we combined ICI with meta-tyrosine-that counteracts immune-suppressive signals-and a selective inhibitor of p38 pathway that proved to counteract the phenomenon of tumor-immunostimulation. RESULTS: ICI were therapeutically effective against both tumor models (proportionally to their immunogenicity) but only when they faced incipient tumors. In contrast, ICI produced acceleration of large and residual tumors. The combined treatment strongly inhibited the growth of large tumors and it managed to cure 80% of mice with local recurrences and 60% of mice bearing residual metastases. CONCLUSIONS: Tumor enhancement was paradoxically correlated to a weak increase of the antitumor IR suggesting that a weak IR - different from a strong tumor-inhibitory one-may produce stimulation of tumor growth, mimicking the HPD observed in some clinical settings.


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Animais , Progressão da Doença , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Camundongos , Neoplasias/tratamento farmacológico , Tirosina
2.
Front Oncol ; 8: 6, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29435437

RESUMO

Immune-checkpoint inhibitors and antitumor vaccines may produce both tumor-inhibitory and tumor-stimulatory effects on growing tumors depending on the stage of tumor growth at which treatment is initiated. These paradoxical results are not necessarily incompatible with current tumor immunology but they might better be explained assuming the involvement of the phenomenon of tumor immunostimulation. This phenomenon was originally postulated on the basis that the immune response (IR) evoked in Winn tests by strong chemical murine tumors was not linear but biphasic, with strong IR producing inhibition and weak IR inducing stimulation of tumor growth. Herein, we extended those former observations to weak spontaneous murine tumors growing in pre-immunized, immune-competent and immune-depressed mice. Furthermore, we demonstrated that the interaction of specifical T cells and target tumor cells at low stimulatory ratios enhanced the production of chemokines aimed to recruit macrophages at the tumor site, which, upon activation of toll-like receptor 4 and p38 signaling pathways, would recruit and activate more macrophages and other inflammatory cells which would produce growth-stimulating signals leading to an accelerated tumor growth. On this basis, the paradoxical effects achieved by immunological therapies on growing tumors could be explained depending upon where the therapy-induced IR stands on the biphasic IR curve at each stage of tumor growth. At stages where tumor growth was enhanced (medium and large-sized tumors), counteraction of the tumor-immunostimulatory effect with anti-inflammatory strategies or, more efficiently, with selective inhibitors of p38 signaling pathways enabled the otherwise tumor-promoting immunological strategies to produce significant inhibition of tumor growth.

3.
Cancer Res ; 72(5): 1043-50, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22315349

RESUMO

Concomitant tumor resistance (CR) is a phenomenon in which a tumor-bearing host is resistant to the growth of secondary tumor implants and metastasis. Although previous studies indicated that T-cell-dependent processes mediate CR in hosts bearing immunogenic small tumors, manifestations of CR induced by immunogenic and nonimmunogenic large tumors have been associated with an elusive serum factor. In a recently published study, we identified this factor as meta-tyrosine and ortho-tyrosine, 2 isomers of tyrosine that would not be present in normal proteins. In 3 different murine models of cancer that generate CR, both meta- and ortho-tyrosine inhibited tumor growth. Additionally, we showed that both isoforms of tyrosine blocked metastasis in a fourth model that does not generate CR but is sensitive to CR induced by other tumors. Mechanistic studies showed that the antitumor effects of the tyrosine isomers were mediated in part by early inhibition of the MAP/ERK pathway and inactivation of STAT3, potentially driving tumor cells into a state of dormancy in G(0)-phase. Other mechanisms, putatively involving the activation of an intra-S-phase checkpoint, would also inhibit tumor proliferation by accumulating cells in S-phase. By revealing a molecular basis for the classical phenomenon of CR, our findings may stimulate new generalized approaches to limit the development of metastases that arise after resection of primary tumors or after other stressors that may promote the escape of metastases from dormancy, an issue that is of pivotal importance to oncologists and their patients.


Assuntos
Neoplasias Pulmonares/secundário , Metástase Neoplásica/imunologia , Neoplasias/patologia , Neoplasias/cirurgia , Tirosina/fisiologia , Animais , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Humanos , Camundongos , Metástase Neoplásica/prevenção & controle , Fase S , Fator de Transcrição STAT3/fisiologia
4.
Cancer Res ; 71(22): 7113-24, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22084446

RESUMO

Concomitant tumor resistance (CR) is a phenomenon originally described in 1906 in which a tumor-bearing host is resistant to the growth of secondary tumor implants and metastasis. Although recent studies have indicated that T-cell-dependent processes mediate CR in hosts bearing immunogenic small tumors, manifestations of CR induced by immunogenic and nonimmunogenic large tumors have been associated with an elusive serum factor. In this study, we identify this serum factor as tyrosine in its meta and ortho isoforms. In three different murine models of cancer that generate CR, both meta-tyrosine and ortho-tyrosine inhibited tumor growth. In addition, we showed that both isoforms of tyrosine blocked metastasis in a fourth model that does not generate CR but is sensitive to CR induced by other tumors. Mechanistic studies showed that the antitumor effects of the tyrosine isoforms were mediated, in part, by early inhibition of mitogen-activated protein/extracellular signal-regulated kinase pathway and inactivation of STAT3, potentially driving tumor cells into a state of dormancy. By revealing a molecular basis for the classical phenomenon of CR, our findings may stimulate new generalized approaches to limit the development of metastases that arise after resection of primary tumors, an issue of pivotal importance to oncologists and their patients.


Assuntos
Transplante de Neoplasias/imunologia , Neoplasias Experimentais/imunologia , Tirosina/fisiologia , Animais , Cromatografia Líquida de Alta Pressão , Resistência à Doença , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Feminino , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/patologia , Fenilalanina/farmacologia , Fator de Transcrição STAT3/fisiologia
5.
Medicina (B Aires) ; 68(4): 301-4, 2008.
Artigo em Espanhol | MEDLINE | ID: mdl-18786887

RESUMO

Up to date, most attempts to use immunotherapy to cause the regression of animal and human established tumors have not been successful. Former experiments have postulated that this failure could be attributed, at least in part, to a lack of immunogenicity of spontaneous tumors. In this paper, we have investigated whether this lack of immunogenicity can be attributed to the absence of tumor antigens or to the existence of tolerogenic mechanisms preventing such antigens from initiating an antitumor immune response. We have used two murine tumors a non-immunogenic spontaneous lymphoma (LB) and a strongly immunogenic methylcholanthrene-induced fibrosarcoma (MC-C) together with a vaccination strategy based on the inoculation of dendritic cells (DC) loaded with a tumor lysate. When DC were pulsed with LB lysate (DC+LB), no maturation of DC was achieved in vitro and no protection against LB implants after DC+LB inoculation was observed in vivo. On the other hand, when DC were pulsed with MC-C lysate (DC+MC-C), maturation of DC was observed along with a strong protection against MC-C implants after DC+MC-C inoculaton. Finally, when DC were pulsed with both LB and MC-C lysates (DC+LB+MC-C), maturation of DC and protection against LB implants were achieved. Since no immune cross reaction between MC-C and LB was ever observed, the most likely interpretation is that LB bears specific tumor antigens but lacks other signals to achieve DC maturation. These signals would be provided by MC-C which would enable DC to mature and to initiate an effective anti-LB immune response.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Fibrossarcoma/imunologia , Linfoma de Células B/imunologia , Animais , Biomarcadores Tumorais , Vacinas Anticâncer/uso terapêutico , Modelos Animais de Doenças , Fibrossarcoma/induzido quimicamente , Imunoterapia/métodos , Linfoma de Células B/induzido quimicamente , Camundongos , Camundongos Endogâmicos BALB C
6.
Clin Med Oncol ; 2: 237-45, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-21892285

RESUMO

Numerous immunization trials have proved successful in preventing the growth of experimental animal tumors and human hepatocarcinomas induced by hepatitis B virus. These results have prompted researchers and physicians to use vaccines in a therapeutic mode but the results have, in general, been disappointing even when strongly immunogenic murine tumors were concerned. Data presented herein suggest that immunotherapy induced by a single dose of a dendritic cell-based vaccine against a murine established tumor or against residual tumor cells after debulking the primary tumor, can render not only inhibitory or null but also stimulatory effects on tumor growth. These different effects might be dependent on where the system is located in the immune response curve that relates the quantity of the immune response to the quantity of target tumor cells. We suggest that high ratios render tumor inhibition, medium and very low ratios render null effects and low ratios-between medium and very low ones-render tumor stimulation. Since the magnitude of these ratios would depend on the antigenic profile of the tumor, the immunogenic strength of the vaccine used and the immunological state of the host, studies aimed to determine the magnitude of these variables in each particular case, seem to be necessary as a pre-condition to design rational immunotherapeutic approaches to cancer. In contrast, if these studies are neglected, the worst thing that an immunotherapist could face is not merely a null effect but enhancement of tumor growth.

7.
Cancer Immunol Immunother ; 57(5): 701-18, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17962945

RESUMO

Although animals can be immunized against the growth of some tumor implants, most of the attempts to use immunotherapy to cause the regression of animal and human tumors once they have become established have been disappointing even when strongly immunogenic tumors were used as target. In this paper, we demonstrate that the failure to achieve an efficient immunological treatment against an established strongly immunogenic murine fibrosarcoma was paralleled with the emergence of a state of immunological unresponsiveness (immunological eclipse) against tumor antigens observed when the tumor surpassed the critical size of 500 mm(3). In turn, the onset of the immunological eclipse was coincidental with the onset of a systemic inflammatory condition characterized by a high number of circulating and splenic polymorphonucleated neutrophils (PMN) displaying activation and Gr1(+)Mac1(+) phenotype and an increasing serum concentration of the pro-inflammatory cytokines TNF-alpha, IL-1beta and IL-6 cytokines and C-reactive protein (CRP) and serum A amyloid (SAA) phase acute proteins. Treatment of tumor-bearing mice with a single low dose (0.75 mg/kg) of the synthetic corticoid dexamethasone (DX) significantly reduced all the systemic inflammatory parameters and simultaneously reversed the immunological eclipse, as evidenced by the restoration of specific T-cell-dependent concomitant immunity, ability of spleen cells to transfer anti-tumor activity and recovery of T-cell signal transduction molecules. Two other anti-inflammatory treatments by using indomethacin or dimeric TNF-alpha receptor, also partially reversed the immunological eclipse although the effect was not as striking as that observed with DX. The reversion of the immunological eclipse was not enough on its own to inhibit the primary growing tumor. However, when we used the two-step strategy of inoculating DX to reverse the eclipse and then dendritic cells loaded with tumor antigens (DC) as an immunization booster, a significant inhibition of the growth of both established tumors and remnant tumor cells after excision of large established tumors was observed, despite the fact that the vaccination alone (DC) had no effect or even enhanced tumor growth in certain circumstances. The two-step strategy of tumor immunotherapy that we present is based on the rationale that it is necessary to eliminate or ameliorate the immunological eclipse as a precondition to allow an otherwise ineffective anti-tumor immunological therapy to have a chance to be successful.


Assuntos
Anti-Inflamatórios/uso terapêutico , Células Dendríticas/transplante , Tolerância Imunológica/efeitos dos fármacos , Imunoterapia/métodos , Inflamação/tratamento farmacológico , Neoplasias Experimentais/terapia , Animais , Western Blotting , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Dexametasona/uso terapêutico , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/imunologia
8.
Medicina (B Aires) ; 67(5): 469-74, 2007.
Artigo em Espanhol | MEDLINE | ID: mdl-18051231

RESUMO

The link between cancer and inflammation in an organ or tissue has firmly been established on the basis that cancer tends to occur at sites of chronic inflammation and that local inflammatory processes can accelerate the growth of preexisting tumors in both animals and human beings. In contrast, the relationship between cancer and systemic inflammation has been less studied. In this work, we demonstrated that the growth of the murine fibrosarcoma MC-C, was accompanied by manifestations of systemic inflammation, as demonstrated by an increase in both the number of circulating polymorphonuclear neutrophils (PMN) and the serum concentration of the proinflammatory cytokines interleukin-1beta (IL-1beta), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha) and the acute phase proteins C reactive (CRP) and serum A amyloid (SAA). Two temporally separate peaks of systemic inflammation were detected during tumor development. The first was displayed during the first week after tumor inoculation. The second peak began around day 14 and its intensity was proportional to tumor size. In mice bearing a large MC-C tumor, a high number of circulating PMN and myeloid precursors were evident. Most of these cells exhibited activation evidenced by an increased reactive oxygen species generation and high expression of the Gr1+/Mac1+ markers. Inoculation of thioglycolate -which generates a transient systemic inflammation-accelerated the growth of MC-C tumor and reciprocally, inhibition of such systemic inflammation by using indomethacin, prevented that enhancing effect. This suggests that the systemic inflammation that the tumor generates on its own, could be part of its growth strategy.


Assuntos
Biomarcadores Tumorais/sangue , Citocinas/sangue , Fibrossarcoma/patologia , Inflamação/patologia , Neoplasias Experimentais/patologia , Animais , Fibrossarcoma/sangue , Inflamação/sangue , Interleucina-1beta/sangue , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/sangue , Proteína Amiloide A Sérica/análise , Fator de Necrose Tumoral alfa/sangue
9.
Medicina (B.Aires) ; 67(5): 469-474, sep.-oct. 2007. graf
Artigo em Espanhol | LILACS | ID: lil-489370

RESUMO

La asociación entre cáncer e inflamación en un órgano o tejido se encuentra sólidamente establecida. En efecto, se sabe que en sitios de inflamación crónica, existe una mayor probabilidad de que se origine un tumor y que procesos inflamatorios locales pueden acelerar el crecimiento de tumores preexistentes en animales y seres humanos. Por otro lado, la relación entre cáncer e inflamación sistémica ha sido menos estudiada. En este trabajo, demostramos que el crecimiento de un fibrosarcoma de ratón (MC-C) fue acompañado por inflamación sistémica, evidenciada por neutrofilia y por un aumento de la concentración sérica de las citoquinas pro-inflamatorias interleuquina-1 beta (IL-1 beta), interleuquina-6 (IL-6) y factor de necrosis tumoral-alfa (TNF-alfa) y de las proteínas de fase aguda C reactiva (CRP) y A amieloide (SAA). Hubo un pico de estas moléculas poco después de la inoculación del tumor, que cayó a valores normales después de la primera semana, para luego comenzar a incrementarse progresivamente en función del tamaño tumoral. Una variación similar fue vista en el porcentaje de neutrófilos polimorfonucleares (PMN) circulantes. En ratones portadores de tumores grandes la mayoría de los PMN exhibían activación evidenciada por aumento en la generación de especies reactivas del oxígeno y alta expresión de los marcadores Gr1+/Mac1+. La inoculación de tioglicolato, que produce una inflamación sistémica transitoria, aceleró el crecimiento de MC-C, mientras que el tratamiento anti-inflamatorio con indometacina revirtió ese efecto. Esto sugiere que MC-C podría utilizar el fenómeno de inflamación sistémica que genera por sí mismo, como parte de su estrategia de crecimiento.


The link between cancer and inflammation in an organ or tissue has firmly been established on the basis that cancer tends to occur at sites of chronic inflammation and that local inflammatory processes can accelerate the growth of preexisting tumors in both animals and human beings. In contrast, the relationship between cancer and systemic inflammation has been less studied. In this work, we demonstrated that the growth of the murine fibrosarcoma MC-C, was accompanied by manifestations of systemic inflammation, as demonstrated by an increase in both the number of circulating polymorphonuclear neutrophils (PMN) and the serum concentration of the proinflammatory cytokines interleukin-1 beta (IL-1 beta), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha) and the acute phase proteins C reactive (CRP) and serum A amyloid (SAA). Two temporally separate peaks of systemic inflammation were detected during tumor development. The first was displayed during the first week after tumor inoculation. The second peak began around day 14 and its intensity was proportional to tumor size. In mice bearing a large MC-C tumor, a high number of circulating PMN and myeloid precursors were evident. Most of these cells exhibited activation evidenced by an increased reactive oxygen species generation and high expression of the Gr1+/Mac1+ markers. Inoculation of thioglycolate -which generates a transient systemic inflammation- accelerated the growth of MC-C tumor and reciprocally, inhibition of such systemic inflammation by using indomethacin, prevented that enhancing effect. This suggests that the systemic inflammation that the tumor generates on its own, could be part of its growth strategy.


Assuntos
Animais , Camundongos , Citocinas/sangue , Fibrossarcoma/patologia , Inflamação/patologia , Neoplasias Experimentais/fisiopatologia , Fibrossarcoma/sangue , Fibrossarcoma/fisiopatologia , Inflamação/sangue , Inflamação/fisiopatologia , Interleucina-1beta/sangue , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/sangue , Proteína Amiloide A Sérica/análise , Biomarcadores Tumorais/sangue , Fator de Necrose Tumoral alfa/sangue
10.
Medicina (B Aires) ; 67(1): 44-8, 2007.
Artigo em Espanhol | MEDLINE | ID: mdl-17408020

RESUMO

Although animals can be prophylactically immunized against the growth of tumor implants, most of the attempts to use immunotherapy to cause the regression of animal and human tumors once they become established have been unsuccessful. To understand the nature of this refractoriness we have studied a methylcholanthrene-induced and strongly immunogenic murine fibrosarcoma. In our model, the onset of this refractoriness was associated with the beginning of an immunosuppressive state known as "immunological eclipse" characterized by a loss of the antitumor immune response when tumor grows beyond a critical size. This immunological eclipse was accompanied by the emergence of a systemic inflammatory condition. Treatment of tumor-bearing mice with a single dose of a synthetic corticosteroid, dexamethasone (DX), reduced significantly all parameters of systemic inflammation and simultaneously reversed the immunological eclipse. The reversion of the eclipse upon DX treatment was not curative itself, but allowed an immunological therapy based in dendritic cells pulsed with tumor antigens, which was itself absolutely ineffective, to exert a significant inhibitory effect against an established growing tumor. The two-step schedule using an anti-inflammatory treatment to reverse the immunological eclipse plus a dendritic cell-based vaccination strategy aimed to stimulate the antitumor immune response, could serve eventually as a model of immunotherapy against animal and human tumors.


Assuntos
Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Fibrossarcoma/tratamento farmacológico , Tolerância Imunológica/imunologia , Sarcoma Experimental/tratamento farmacológico , Animais , Anti-Inflamatórios/imunologia , Anti-Inflamatórios/uso terapêutico , Vacinas Anticâncer/imunologia , Carcinógenos , Dexametasona/imunologia , Dexametasona/uso terapêutico , Intervalo Livre de Doença , Fibrossarcoma/induzido quimicamente , Fibrossarcoma/imunologia , Tolerância Imunológica/efeitos dos fármacos , Inflamação/tratamento farmacológico , Metilcolantreno , Camundongos , Camundongos Endogâmicos BALB C , Sarcoma Experimental/induzido quimicamente , Sarcoma Experimental/imunologia
11.
Medicina (B.Aires) ; 67(1): 44-48, jan.-fev. 2007. graf
Artigo em Espanhol | BINACIS | ID: bin-123134

RESUMO

Aunque existen vacunas para prevenir la aparición de tumores en animales de experimentación, la mayoría de los intentos por aplicar aquellas vacunas con fines terapéuticos contra tumores establecidos no han sido exitosos. Para comprender la naturaleza de esta refractariedad, estudiamos un tumor de ratón fuertemente inmunogénico inducido por el carcinógeno químico metilcolantreno. En nuestro modelo, el inicio de esta refractariedad coincidió con el comienzo de un estado de inmunosupresión conocido como "eclipse inmunológico" caracterizado por una pérdida o bloqueo de la respuesta inmune antitumoral después que el tumor ha superado cierto tamaño crítico. Este eclipse inmunológico fue acompañado por un proceso de inflamación sistémica en el organismo. El tratamiento de los ratones portadores de tumor con una única dosis del corticoide sintético dexametasona (DX) redujo los parámetros de inflamación sistémica e indujo la reversión del eclipse. Esta reversión no fue por sí misma curativa pero permitió que un tratamiento inmunológico basado en células dendríticas estimuladas con antígenos tumorales, que por sí solo era absolutamente ineficaz, pudiera ejercer un significativo efecto inhibidor sobre un tumor en crecimiento. El esquema de dos pasos que comprende, primero, un tratamiento antiinflamatorio para revertir el eclipse y segundo, una estrategia de vacunación basada en células dendríticas destinada a estimular la respuesta inmune antitumoral, podría servir, eventualmente, como un modelo de inmunoterapia contra tumores en animales y seres humanos.(AU)


Although animals can be prophylactically immunized against the growth of tumor implants, most of the attempts to use immunotherapy to cause the regression of animal and human tumors once they become established have been unsuccessful. To understand the nature of this refractoriness we have studied a methylcholanthrene-induced and strongly immunogenic murine fibrosarcoma. In our model, the onset of this refractoriness was associated with the beginning of an immunosuppressive state known as "immunological eclipse" characterized by a loss of the antitumor immune response when tumor grows beyond a critical size. This immunological eclipse was accompanied by the emergence of a systemic inflammatory condition. Treatment of tumor-bearing mice with a single dose of a synthetic corticosteroid, dexamethasone (DX), reduced significantly all parameters of systemic inflammation and simultaneously reversed the immunological eclipse. The reversion of the eclipse upon DX treatment was not curative itself, but allowed an immunological therapy based in dendritic cells pulsed with tumor antigens, which was itself absolutely ineffective, to exert a significant inhibitory effect against an established growing tumor. The two-step schedule using an anti-inflammatory treatment to reverse the immunological eclipse plus a dendritic cell-based vaccination strategy aimed to stimulate the anti-tumor immune response, could serve eventually as a model of immunotherapy against animal and human tumors. (AU)


Assuntos
Humanos , Animais , Camundongos , Vacinas Anticâncer/uso terapêutico , Fibrossarcoma/tratamento farmacológico , Sarcoma Experimental/tratamento farmacológico , Terapia de Imunossupressão/métodos , Células Dendríticas/imunologia , Dexametasona/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Vacinas Anticâncer/imunologia , Fibrossarcoma/imunologia , Sarcoma Experimental/imunologia , Dexametasona/imunologia , Anti-Inflamatórios/imunologia , Camundongos Endogâmicos BALB C , Metilcolantreno/efeitos adversos , Inflamação/tratamento farmacológico , Intervalo Livre de Doença
12.
Medicina (B.Aires) ; 67(1): 44-48, jan.-fev. 2007. graf
Artigo em Espanhol | LILACS | ID: lil-464743

RESUMO

Aunque existen vacunas para prevenir la aparición de tumores en animales de experimentación, la mayoría de los intentos por aplicar aquellas vacunas con fines terapéuticos contra tumores establecidos no han sido exitosos. Para comprender la naturaleza de esta refractariedad, estudiamos un tumor de ratón fuertemente inmunogénico inducido por el carcinógeno químico metilcolantreno. En nuestro modelo, el inicio de esta refractariedad coincidió con el comienzo de un estado de inmunosupresión conocido como "eclipse inmunológico" caracterizado por una pérdida o bloqueo de la respuesta inmune antitumoral después que el tumor ha superado cierto tamaño crítico. Este eclipse inmunológico fue acompañado por un proceso de inflamación sistémica en el organismo. El tratamiento de los ratones portadores de tumor con una única dosis del corticoide sintético dexametasona (DX) redujo los parámetros de inflamación sistémica e indujo la reversión del eclipse. Esta reversión no fue por sí misma curativa pero permitió que un tratamiento inmunológico basado en células dendríticas estimuladas con antígenos tumorales, que por sí solo era absolutamente ineficaz, pudiera ejercer un significativo efecto inhibidor sobre un tumor en crecimiento. El esquema de dos pasos que comprende, primero, un tratamiento antiinflamatorio para revertir el eclipse y segundo, una estrategia de vacunación basada en células dendríticas destinada a estimular la respuesta inmune antitumoral, podría servir, eventualmente, como un modelo de inmunoterapia contra tumores en animales y seres humanos.


Although animals can be prophylactically immunized against the growth of tumor implants, most of the attempts to use immunotherapy to cause the regression of animal and human tumors once they become established have been unsuccessful. To understand the nature of this refractoriness we have studied a methylcholanthrene-induced and strongly immunogenic murine fibrosarcoma. In our model, the onset of this refractoriness was associated with the beginning of an immunosuppressive state known as "immunological eclipse" characterized by a loss of the antitumor immune response when tumor grows beyond a critical size. This immunological eclipse was accompanied by the emergence of a systemic inflammatory condition. Treatment of tumor-bearing mice with a single dose of a synthetic corticosteroid, dexamethasone (DX), reduced significantly all parameters of systemic inflammation and simultaneously reversed the immunological eclipse. The reversion of the eclipse upon DX treatment was not curative itself, but allowed an immunological therapy based in dendritic cells pulsed with tumor antigens, which was itself absolutely ineffective, to exert a significant inhibitory effect against an established growing tumor. The two-step schedule using an anti-inflammatory treatment to reverse the immunological eclipse plus a dendritic cell-based vaccination strategy aimed to stimulate the anti-tumor immune response, could serve eventually as a model of immunotherapy against animal and human tumors.


Assuntos
Humanos , Animais , Camundongos , Anti-Inflamatórios/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Dexametasona/uso terapêutico , Fibrossarcoma/tratamento farmacológico , Terapia de Imunossupressão/métodos , Sarcoma Experimental/tratamento farmacológico , Anti-Inflamatórios/imunologia , Vacinas Anticâncer/imunologia , Intervalo Livre de Doença , Dexametasona/imunologia , Fibrossarcoma/imunologia , Inflamação/tratamento farmacológico , Camundongos Endogâmicos BALB C , Metilcolantreno/efeitos adversos , Sarcoma Experimental/imunologia
13.
Theor Biol Med Model ; 3: 43, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17173673

RESUMO

BACKGROUND: Most theories about cancer proposed during the last century share a common denominator: cancer is believed to be a biological nonsense for the organism in which it originates, since cancer cells are believed to be ones evading the rules that control normal cell proliferation and differentiation. In this essay, we have challenged this interpretation on the basis that, throughout the animal kingdom, cancer seems to arise only in injured organs and tissues that display lost or diminished regenerative ability. HYPOTHESIS: According to our hypothesis, a tumor cell would be the only one able to respond to the demand to proliferate in the organ of origin. It would be surrounded by "normal" aged cells that cannot respond to that signal. According to this interpretation, cancer would have a profound biological sense: it would be the ultimate way to attempt to restore organ functions and structures that have been lost or altered by aging or noxious environmental agents. In this way, the features commonly associated with tumor cells could be reinterpreted as progressively acquired adaptations for responding to a permanent regenerative signal in the context of tissue injury. Analogously, several embryo developmental stages could be dependent on cellular damage and death, which together disrupt the field topography. However, unlike normal structures, cancer would have no physiological value, because the usually poor or non-functional nature of its cells would make their reparative task unattainable. CONCLUSION: The hypothesis advanced in this essay might have significant practical implications. All conventional therapies against cancer attempt to kill all cancer cells. However, according to our hypothesis, the problem might not be solved even if all the tumor cells were eradicated. In effect, if the organ failure remained, new tumor cells would emerge and the tumor would reinitiate its progressive growth in response to the permanent regenerative signal of the non-restored organ. Therefore, efficient anti-cancer therapy should combine an attack against the tumor cells themselves with the correction of the organ failure, which, according to this hypothesis, is fundamental to the origin of the cancer.


Assuntos
Diferenciação Celular , Proliferação de Células , Neoplasias/fisiopatologia , Adaptação Fisiológica , Senescência Celular , Desenvolvimento Embrionário , Humanos , Neoplasias/terapia
14.
Pediatr Nephrol ; 21(8): 1082-92, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16703367

RESUMO

Uninephrectomy (Unx) is followed by the compensatory renal growth (CRG) of the remaining kidney. Previous evidence has shown that during CRG, renal tissue is resistant to a variety of pathologies. We tested the hypothesis that the functional changes that take place during CRG could attenuate Shiga toxin (Stx) toxicity in a mouse model of Stx2-induced hemolytic uremic syndrome (HUS). The participation of nitric oxide (NO) was analyzed. After CRG induction with Unx, mice were exposed to a lethal dose of Stx2, and the degree of renal damage and mortality was measured. Stx2 effects on the growth, renal blood flow (RBF) and NO synthase (NOS) intrarenal expression in the remaining kidney were then studied. The induction of CRG strongly prevented Stx2-mediated mortality and renal damage. Administration of the NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME) during CRG partially impaired the protection. Both Stx2 and L-NAME interfered with the hypertrophic and hyperplastic responses to Unx, as well as with the increase in RBF. In intact mice, Stx2 decreased renal perfusion, inhibited endothelial NOS basal expression and enhanced inducible NOS expression; all of these effects were attenuated by prior Unx. It is concluded that during CRG mice are highly protected against Stx2 toxicity and lethality. The protective capacity of CRG could be related to the enhancement of renal perfusion and preservation of eNOS renal expression, counterbalancing two major pathogenic mechanisms of Stx2.


Assuntos
Síndrome Hemolítico-Urêmica/induzido quimicamente , Síndrome Hemolítico-Urêmica/prevenção & controle , Rim/crescimento & desenvolvimento , Toxina Shiga II/toxicidade , Animais , Síndrome Hemolítico-Urêmica/enzimologia , Masculino , Camundongos , Óxido Nítrico/fisiologia
15.
Oncol Res ; 15(3): 169-82, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16050138

RESUMO

A small primary or secondary tumor load can occasionally induce more deleterious effects than a histologically identical larger one. In the four murine models studied herein this enhanced tumor aggressiveness could not be attributed to NRAS mutations or other hereditary changes, differential vascularization of live tumor tissues, or necrosis content. Instead, the main tumor feature associated with a more aggressive behavior was the presence of a high number of vessels, sometimes filled with inflammatory cells, inside a tumor area, which we have identified and designated as the transition zone between the live and the necrotic zones. Our experiments suggest that during tumor growth, different cachectic factors are produced within the transition and necrotic zones by dying tumor cells and by tumor infiltrating macrophages only reaching the general circulation through the vessels present in the transition zone. Therefore, a small tumor displaying high vascularization of its transition area could be harmful to its host, while, in contrast, a large tumor could behave as a relatively benign one if its transition zone exhibited little or no vascularization, and in consequence its cachectic factors remained "trapped." Similar histological images to those observed in mice were seen in a significant percentage of human cancer biopsies, raising the possibility that such images might have a prognostic value.


Assuntos
Neoplasias/patologia , Neovascularização Patológica/patologia , Animais , Biomarcadores Tumorais/análise , Linhagem Celular Tumoral , DNA de Neoplasias/análise , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Necrose , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias/irrigação sanguínea , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas/genética , RNA Neoplásico/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Oncol Res ; 13(3): 147-60, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12549624

RESUMO

A differential effect of pregnancy on the growth of subcutaneous implants of four murine tumors has been observed. Two tumors lacking receptors for progesterone and estrogen [methylcholanthrene-induced fibrosarcoma (MC-C) and spontaneous lymphoid leukemia (LB)] exhibited slow kinetics throughout the course of pregnancy, although inhibition was stronger beyond day 10. On the other hand, one of two tumors bearing receptors for progesterone and estrogen [medroxyprogesterone (MPA)-induced mammary adenocarcinoma (C7HI)] exhibited three phases: up to days 8-10 of gestation the tumor grew faster than in virgins, between days 8-10 and 15 it reached a plateau, and beyond day 15 a sharp reduction in tumor mass was observed. The other tumor [mouse mammary tumor virus (MMTV)-induced mammary carcinoma(T2280)] behaved as a typical pregnancy-dependent tumor (i.e., it grew in pregnant but not in virgin mice, regressed soon after delivery, and reassumed its growth at the middle of a second round of pregnancy). Neither MPA nor estrogen affected MC-C and LB tumor growth. On the other hand, MPA-treated mice enhanced C7HI tumor and reciprocally C7HI tumor-bearing mice treated with estrogen strongly inhibited tumor growth. As for T2280, neither MPA nor estrogen alone could promote tumor growth and, in consequence, no tumor developed. However, when MPA plus estrogen was administered in a schedule simulating the successive appearance of these hormones in pregnancy, T2280 grew even faster than in pregnant mice. When the four tumors were implanted in mice bearing grafts of embryonal tissues (teratomas), all of them were inhibited. This antitumor effect was similar to that observed in pregnancy when tumors unresponsive to progesterone and estrogen were tested. On the other hand, with tumors bearing progesterone and estrogen receptors, differences in tumor growth were detected in pregnant and teratoma-bearing mice. This suggested the existence during pregnancy of two factors potentially acting on tumor growth. First, a progesterone and estrogen-mediated hormonal component, which would exert either inhibitory or stimulatory effects only evidenced with tumors bearing hormonal receptors. Secondly, an antitumor effect proportional to the growing embryonal mass, inhibiting all tumors independently of their origin or hormone responsiveness. This antitumor effect could be attributed to a beat-resistant serum factor (1,000-1,200 Da molecular weight) presumably associated with the pathway of the arachidonic acid metabolism. The interplay between the hormonal component and the serum factor associated with embryonal mass could account for some of the largely heterogeneous and otherwise unexplained effects of pregnancy on tumor growth reported in the literature and illustrated by the four tumors studied here.


Assuntos
Hormônios/fisiologia , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/fisiopatologia , Complicações Neoplásicas na Gravidez/fisiopatologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Adenocarcinoma/fisiopatologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Estrogênios/farmacologia , Feminino , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/patologia , Fibrossarcoma/fisiopatologia , Hormônios/farmacologia , Indometacina/farmacologia , Leucemia Linfoide/tratamento farmacológico , Leucemia Linfoide/patologia , Leucemia Linfoide/fisiopatologia , Masculino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Neovascularização Patológica , Gravidez , Complicações Neoplásicas na Gravidez/patologia , Progesterona/metabolismo , Progesterona/farmacologia , Receptores de Progesterona/metabolismo , Teratoma/patologia , Teratoma/fisiopatologia
17.
Bol. Acad. Nac. Med. B.Aires ; 75(1): 145-52, jun. 1997. tab, graf
Artigo em Espanhol | BINACIS | ID: bin-18179

RESUMO

ß-Lapachona prolonga la sobrevida de ratones con leucemia inducida por el virus de Friend y de pollos con sarcoma inducido por el virus de Rous y recientemente ha demostrado inhibir la proliferación in vitro de varias líneas tumorales humanas. La estructura química de esta droga y presumiblemente su modo de acción, son muy diferentes de la estructura y modo de acción de las drogas antitumorales comúnmente utilizadas en la terapia. Por tal motivo, y con el propósito de ampliar el conocimiento sobre nuevas drogas antineoplásicas intentamos determinar, en este trabajo, si ß-lapachona era capaz de inhibir in vivo el desarrollo de un tumor de ratón no inducido por virus que ha demostrado ser refractario al tratamiento con drogas antineoplásicas convencionales. Se mostró que la administración oral y diaria de ß-lapachona redujo la toma tumoral y prolongó la sobrevida en aquellos ratones donde el tumor LB había crecido. El mayor efecto se produjo cuando la droga fue administrada en la concentración de 125 mg/Kg/día simultáneamente con un inóculo tumoral de 10n ó 10 elevado a la 4, células LB. El efecto sobre un inóculo mayor (10 elevado a la 5, células) o sobre un tumor vascularizado y en activo crecimiento fue más tenue aunque igualmente perceptible. Estos resultados son, en principio, promisorios, pero más experimentos, con diferentes concentraciones de la droga y con diferentes modelos tumorales serán necesarios para determinar adecuadamente el valor terapéutico de ß-lapachona sobre tumores murinos.(AU)


Assuntos
Animais , Camundongos , Naftoquinonas/uso terapêutico , Naftoquinonas/administração & dosagem , Antineoplásicos , Sobreviventes , Vírus da Leucemia Murina de Friend , Leucemia
18.
Bol. Acad. Nac. Med. B.Aires ; 75(1): 145-52, jun. 1997. tab, graf
Artigo em Espanhol | LILACS | ID: lil-216250

RESUMO

ß-Lapachona prolonga la sobrevida de ratones con leucemia inducida por el virus de Friend y de pollos con sarcoma inducido por el virus de Rous y recientemente ha demostrado inhibir la proliferación in vitro de varias líneas tumorales humanas. La estructura química de esta droga y presumiblemente su modo de acción, son muy diferentes de la estructura y modo de acción de las drogas antitumorales comúnmente utilizadas en la terapia. Por tal motivo, y con el propósito de ampliar el conocimiento sobre nuevas drogas antineoplásicas intentamos determinar, en este trabajo, si ß-lapachona era capaz de inhibir in vivo el desarrollo de un tumor de ratón no inducido por virus que ha demostrado ser refractario al tratamiento con drogas antineoplásicas convencionales. Se mostró que la administración oral y diaria de ß-lapachona redujo la toma tumoral y prolongó la sobrevida en aquellos ratones donde el tumor LB había crecido. El mayor efecto se produjo cuando la droga fue administrada en la concentración de 125 mg/Kg/día simultáneamente con un inóculo tumoral de 10n ó 10 elevado a la 4, células LB. El efecto sobre un inóculo mayor (10 elevado a la 5, células) o sobre un tumor vascularizado y en activo crecimiento fue más tenue aunque igualmente perceptible. Estos resultados son, en principio, promisorios, pero más experimentos, con diferentes concentraciones de la droga y con diferentes modelos tumorales serán necesarios para determinar adecuadamente el valor terapéutico de ß-lapachona sobre tumores murinos.


Assuntos
Animais , Camundongos , Antineoplásicos , Vírus da Leucemia Murina de Friend , Leucemia , Naftoquinonas , Naftoquinonas/uso terapêutico , Sobreviventes
19.
Buenos Aires; s.n; 1997. 60 p. ilus, graf.
Monografia em Espanhol | BINACIS | ID: biblio-1205423

RESUMO

La resistencia concomitante antitumoral es el fenómeno por el cual un individuo portador de un tumor inhibe el crecimiento de un implante tumoral secundario realizado en un sitio distante del primero. En este trabajo hemos evaluado este fenómeno en ratones eutímicos y atímicos usando 9 tumores con diferente grado de inmunogenicidad. Durante el desarrollo tumoral, se observaron dos picos, temporalmente separados de RC: el primer pico fue detectado sólo con tumores inmunogénicos pequeños (menos de 500 mm3); fue específico de tumor y mediado por mecanismos inmunológicos clásicos dependientes del timo. El segundo pico fue exhibido por tumores grandes (más de 2000 mm3) independientemente y correlacionó con la presencia de un factor(es) del suero (ni anticuerpos ni complemento) que inhibía in vitro la proliferación de las células tumorales... (TRUNCADO)


Assuntos
Animais , Camundongos , Aminoácidos/análise , Apoptose , Citometria de Fluxo , Cromatografia , Cromatografia Gasosa-Espectrometria de Massas , Hidrólise , Imuno-Histoquímica , Inibidores do Crescimento , Metástase Neoplásica/imunologia , Neoplasias/imunologia
20.
Buenos Aires; s.n; 1997. 60 p. ilus, graf. (83467).
Monografia em Espanhol | BINACIS | ID: bin-83467

RESUMO

La resistencia concomitante antitumoral es el fenómeno por el cual un individuo portador de un tumor inhibe el crecimiento de un implante tumoral secundario realizado en un sitio distante del primero. En este trabajo hemos evaluado este fenómeno en ratones eutímicos y atímicos usando 9 tumores con diferente grado de inmunogenicidad. Durante el desarrollo tumoral, se observaron dos picos, temporalmente separados de RC: el primer pico fue detectado sólo con tumores inmunogénicos pequeños (menos de 500 mm3); fue específico de tumor y mediado por mecanismos inmunológicos clásicos dependientes del timo. El segundo pico fue exhibido por tumores grandes (más de 2000 mm3) independientemente y correlacionó con la presencia de un factor(es) del suero (ni anticuerpos ni complemento) que inhibía in vitro la proliferación de las células tumorales... (TRUNCADO)(AU)


Assuntos
Animais , Camundongos , Imuno-Histoquímica , Apoptose , Citometria de Fluxo , Cromatografia , Hidrólise , Cromatografia Gasosa-Espectrometria de Massas , Inibidores do Crescimento , Neoplasias/imunologia , Metástase Neoplásica/imunologia , Aminoácidos/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...