Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Nat Med ; 1(8): 786-91, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7585181

RESUMO

Activation of transcription of the Egr-1 gene by X-rays is regulated by the promoter region of this gene. We linked the radiation-inducible promoter region of the Egr-1 gene to the gene encoding the radiosensitizing and tumoricidal cytokine, tumour necrosis factor-alpha (TNF-alpha) and used a replication-deficient adenovirus to deliver the Egr-TNF construct to human tumours growing in nude mice. Combined treatment with Ad5.Egr-TNF and 5,000 cGy (rad) resulted in increased intratumoral TNF-alpha production and increased tumour control compared with treatment with Ad5.Egr-TNF alone or with radiation alone. The increase in tumour control was achieved without an increase in normal tissue damage when compared to tissue injury from radiation alone. Control of gene transcription by ionizing radiation in vivo represents a novel method of spatial and temporal regulation of gene-based medical treatments.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/efeitos da radiação , Terapia Genética/métodos , Proteínas Imediatamente Precoces , Neoplasias Laríngeas/terapia , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/genética , Animais , Apoptose , Proteínas de Ligação a DNA/biossíntese , Proteína 1 de Resposta de Crescimento Precoce , Vetores Genéticos , Humanos , Imuno-Histoquímica , Neoplasias Laríngeas/radioterapia , Mastadenovirus/genética , Camundongos , Camundongos Nus , Necrose , Transplante de Neoplasias , Radiação Ionizante , Proteínas Recombinantes de Fusão , Fatores de Transcrição/biossíntese , Transplante Heterólogo , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/biossíntese
2.
J Natl Cancer Inst ; 45(5): 997-1004, 1970 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18605426

RESUMO

Transplacental hemorrhage is reportedly detectable by erythrocytes containing fetal hemoglobin-F in the maternal circulation. The procedure in the present study is based on the premise that fetal hemoglobin is not eluted from red cells in blood smears by an acid buffer, whereas adult hemoglobin is removed. Erythrocytes containing hemoglobin-F were observed in blood smears of both virgin and pregnant BALB/c mice, but the relative number of these cells in the maternal blood increased during pregnancy, reached a peak around the time of parturition, and decreased thereafter. In BALB/c females pregnant by DBA/2 males, the relatively large numbers of acid-resistant erythrocytes in maternal blood were related to the maternal unresponsiveness to DBA/2 allografts. The fetal derivation of at least a fraction of these cells was suggested by the apparent activity of fetal immunogens in the maternal circulation. The blood of peripartum BALB/c mice multiparous by DBA/2 males was injected into normal, nonimmune BALB/c mice. The blood recipients were later challenged with an allotransplantable DBA/2 tumor. A low degree of immunity was indicated by a decreased size of tumor implants. Attempts to demonstrate vertical transmission of immune suppression from mothers to progeny were negative. These findings indicate that transplacental hemorrhage is a potential source of antigens involved in the induction of specific maternal unresponsiveness.


Assuntos
Hemoglobina Fetal/imunologia , Transplante de Neoplasias/imunologia , Doenças Placentárias/imunologia , Hemorragia Uterina/imunologia , Animais , Quimera/imunologia , Eritrócitos/imunologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Paridade , Parto/imunologia , Doenças Placentárias/sangue , Gravidez , Transplante Homólogo , Hemorragia Uterina/sangue
3.
Cancer Res ; 55(23): 5561-5, 1995 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-7585635

RESUMO

We report that radiation enhances gene therapy of a radioresistant tumor by upregulating the induction of a chimeric gene encoding a radiosensitizing protein, tumor necrosis factor alpha (TNF-alpha). We ligated the radiation-inducible CArG elements of the radiation-inducible Egr-1 promoter/enhancer region upstream to the transcriptional start site of the human TNF cDNA (pE425-TNF). This construct was transfected using cationic liposomes into the variant murine fibrosarcoma cell line, P4L. The P4L cell line was both radioresistant (D0 = 188) and resistant to TNF. After a single intratumoral injection of 10 micrograms of pE425-TNF in cationic liposomes and two 20-Gy doses of irradiation, mean tumor volumes were significantly reduced in P4L tumors as compared to those receiving either pE425-TNF in liposomes or radiation alone (P = 0.01). TNf protein in P4L tumors was induced by radiation as high as 29 times control levels and remained detectable for 14 days. Our data indicate that combined gene therapy using liposomes, together with ionizing radiation to locally activate the induction of a radiosensitizing protein, is successful at overcoming resistance to both TNF and radiation.


Assuntos
Fibrossarcoma/terapia , Terapia Genética , Neoplasias Experimentais/terapia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Terapia Combinada/métodos , Feminino , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Fibrossarcoma/radioterapia , Lipossomos , Camundongos , Camundongos Nus , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Experimentais/radioterapia , Neoplasias Induzidas por Radiação , Tolerância a Radiação , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/metabolismo
4.
Cancer Res ; 56(19): 4311-4, 1996 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-8813113

RESUMO

Intratumoral injection of an adenoviral vector containing radiation-inducible DNA sequences of the Egr-1 promoter linked to a cDNA encoding tumor necrosis factor (TNF) alpha (Ad.Egr-TNF) enhances the tumoricidal action of ionizing radiation in a human epidermoid carcinoma xenograft (SQ-20B). The dominant histopathological feature in tumor-bearing animals treated with Ad.Egr-TNF and irradiation is extensive intratumoral vascular thrombosis and tumor necrosis. Thrombosis and necrosis are not observed in animals treated with either the viral construct encoding TNF-alpha or radiation and did not occur in irradiated normal tissues adjacent to tumor in animals injected with Ad.Egr-TNF. To determine if the occlusive effects of Ad.Egr-TNF and X-irradiation were specific for tumor vessels, non-tumor-bearing mice were irradiated after receiving i.m. injection of Ad.Egr-TNF at viral titers 20-100 times greater than titers injected intratumorally. No vascular thrombosis was observed in the treated normal tissues. Combined Ad.Egr-TNF and radiation produced occlusion of tumor microvessels without significant normal tissue damage. Taken together, these data suggest that the interaction between radiation inducible TNF-alpha and X-irradiation occurs selectively within the tumor vessels.


Assuntos
Carcinoma de Células Escamosas/terapia , Regulação da Expressão Gênica/efeitos da radiação , Terapia Genética , Neovascularização Patológica/genética , Fator de Necrose Tumoral alfa/genética , Animais , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/patologia , Feminino , Vetores Genéticos/genética , Vetores Genéticos/efeitos da radiação , Humanos , Camundongos , Camundongos Nus , Necrose , Transplante de Neoplasias , Proteínas Recombinantes de Fusão/biossíntese , Trombose/etiologia , Fator de Necrose Tumoral alfa/biossíntese
5.
Cancer Res ; 57(19): 4205-9, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9331076

RESUMO

Gene therapy combined with radiation therapy to enhance selectively radiation cytotoxicity in malignant cells represents a new approach for cancer treatment. We investigated the efficacy of adenoviral (Ad5)-directed cytosine deaminase/5-fluorocytosine (CD/5-FC) enzyme/prodrug gene therapy to enhance selectively the tumoricidal action of ionizing radiation in human cancer xenografts derived from a human squamous carcinoma cell line (SQ-20B). Tumor xenografts grown in hindlimbs of nude mice were transfected with an adenoviral vector (Ad.CMV.CD) containing the cytosine deaminase (CD) gene under the control of a cytomegalovirus (CMV) promoter. Mice were injected i.p. with 800 mg/kg of 5-FC for 12 days, and tumors were treated with fractionated radiation at a dose of 5 Gy/day to a total dose of 50 Gy. In larger tumors with a mean volume of 1069 mm3, marked tumor regression to 11% of the original tumor volume was observed at day 21 (P = 0.01). The volumetric regression of smaller tumors with a mean volume of 199 mm3, which received the same combined treatment protocol, was significant at day 12 (P = 0.014). However, unlike large tumors, regression of the smaller tumors continued until day 36 (P = 0.01), with 43% cured at day 26. No cures or significant volumetric reduction in size was observed in tumors treated with radiation alone; Ad.CMV.CD with or without radiation; or with Ad.CMV.CD and 5-FC. These results suggest that the CD/5-FC gene therapy approach is an effective radiosensitizing strategy and may lead to substantial improvement in local tumor control that would translate into improved cure rates and better survival.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Flucitosina/uso terapêutico , Terapia Genética , Neoplasias Experimentais/terapia , Nucleosídeo Desaminases/uso terapêutico , Radiossensibilizantes/uso terapêutico , Adenovírus Humanos/genética , Animais , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/radioterapia , Carcinoma de Células Escamosas/terapia , Terapia Combinada , Citosina Desaminase , Feminino , Vetores Genéticos , Humanos , Neoplasias Laríngeas/tratamento farmacológico , Neoplasias Laríngeas/radioterapia , Neoplasias Laríngeas/terapia , Camundongos , Camundongos Nus , Recidiva Local de Neoplasia , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Neoplasias Experimentais/radioterapia , Nucleosídeo Desaminases/genética , Tolerância a Radiação , Proteínas Recombinantes de Fusão/uso terapêutico , Transfecção , Transplante Heterólogo
6.
Cancer Res ; 57(19): 4340-7, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9331096

RESUMO

Approximately 30% of cancer deaths result from the failure to control local and regional tumors. The goal of radiotherapy is to maximize local and regional tumor cell killing while minimizing normal tissue destruction. Attempts to enhance radiation-mediated tumor cell killing using halogenated pyrimidines, antimetabolites, and other DNA-damaging agents or sensitizers of hypoxic tumor cells have met with only modest clinical success. In an unique strategy to modify tumor radiosensitivity, we used an inhibitor of the protein kinase C group A and B isoforms, chelerythrine chloride (chelerythrine), to enhance the killing effects of ionizing radiation (IR). Protein kinase C activity plays a central role in cellular proliferation, differentiation, and apoptosis. Chelerythrine increases sphingomyelinase activity and enhances IR-mediated cell killing through induction of apoptotic tumor cell death in a radioresistant tumor model both in vitro and in vivo. Although previous reports have suggested that IR-mediated apoptosis correlates with tumor volume reduction, we demonstrate for the first time that lowering the apoptotic threshold increases tumor cell killing in vivo.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/radioterapia , Traumatismos Craniocerebrais/radioterapia , Inibidores Enzimáticos/farmacologia , Isoenzimas/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Fenantridinas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Radiossensibilizantes/uso terapêutico , Esfingomielina Fosfodiesterase/metabolismo , Alcaloides , Animais , Benzofenantridinas , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/enzimologia , Ceramidas/farmacologia , Quimioterapia Adjuvante , Terapia Combinada , Traumatismos Craniocerebrais/tratamento farmacológico , Traumatismos Craniocerebrais/enzimologia , Endopeptidases/metabolismo , Ativação Enzimática/efeitos dos fármacos , Isoenzimas/metabolismo , Camundongos , Camundongos Nus , Proteínas de Neoplasias/antagonistas & inibidores , Proteína Quinase C/metabolismo , Radiossensibilizantes/farmacologia , Transplante Heterólogo
7.
Cancer Res ; 54(16): 4266-9, 1994 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8044769

RESUMO

Transcriptional regulation of the promoter/enhancer region of the Egr-1 gene is activated by ionizing radiation. We linked DNA sequences from the promotor region of Egr-1 to a complementary DNA sequence which encodes human tumor necrosis factor (TNF) alpha, a radiosensitizing cytokine. The Egr-TNF construct was transfected into a human cell line of hematopoietic origin, HL525, which was used in an experimental animal system. HL525 (clone 2) cells containing the Egr-TNF construct which exhibits radiation induction of TNF-alpha were injected into human xenografts of the radioresistant human squamous cell carcinoma cell line SQ-20B. Animals treated with radiation and clone 2 demonstrated an increase in tumor cures compared with animals treated with radiation alone or unirradiated animals given injections of clone 2 alone. No increase in local or systemic toxicity was observed in the combined treatment group. The combination of gene therapy and radiation therapy enhances tumor cures without increasing normal tissue toxicity and is a new paradigm for cancer treatment.


Assuntos
Carcinoma de Células Escamosas/terapia , DNA Complementar/genética , Genes Precoces/genética , Terapia Genética/métodos , Leucemia/terapia , Fator de Necrose Tumoral alfa/genética , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/radioterapia , Cloranfenicol O-Acetiltransferase/análise , Cloranfenicol O-Acetiltransferase/genética , Terapia Combinada , Estudos de Viabilidade , Feminino , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/radioterapia , Camundongos , Camundongos Nus , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo
8.
Cancer Res ; 58(24): 5686-9, 1998 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-9865723

RESUMO

Angiostatin, a proteolytic fragment of plasminogen, inhibits the growth of primary and metastatic tumors by suppressing angiogenesis. When used in combination with ionizing radiation (IR), angiostatin demonstrates potent antitumor synergism, largely caused by inhibition of the tumor microvasculature. We report here the temporal interaction of angiostatin and IR in Lewis lung carcinoma (LLC) tumors growing in the hind limbs of syngeneic mice. Tumors with an initial mean volume of 510 +/- 151 mm3 were treated with IR alone (20 Gy x 2 doses on days 0 and 1), angiostatin alone (25 mg/kg/day divided twice daily) on days 0 through 13, or a combination of the two as follows: (a) IR plus angiostatin (days 0 through 13); (b) IR plus angiostatin (days 0 and 1); and (c) IR followed by angiostatin beginning on the day after IR completion and given daily thereafter (days 2 through 13). By day 14, tumors in untreated control mice had grown to 6110 +/- 582 mm3, whereas in mice treated with: (a) IR alone, tumors had grown to 2854 +/- 338 mm3 (P < 0.05 compared with untreated controls); and (b) angiostatin alone, tumors had grown to 3666 +/- 453 mm3 (P < 0.05 compared with untreated controls). In combined-treatment groups, in mice treated with: (a) IR plus longer-course angiostatin, tumors reached 2022 +/- 282 mm3 (P = 0.036 compared with IR alone); (b) IR followed by angiostatin, tumors reached 2677 +/- 469 mm3 (P > 0.05 compared with IR alone); and (c) IR plus short-course angiostatin, tumors reached 1032 +/- 78 mm3 (P < 0.001 compared with IR alone). These findings demonstrate that the efficacy of experimental radiation therapy is potentiated by brief concomitant exposure of the tumor vasculature to angiostatin.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/radioterapia , Fragmentos de Peptídeos/uso terapêutico , Plasminogênio/uso terapêutico , Angiostatinas , Animais , Terapia Combinada , Esquema de Medicação , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/administração & dosagem , Plasminogênio/administração & dosagem , Fatores de Tempo , Células Tumorais Cultivadas
9.
Cancer Res ; 59(14): 3374-8, 1999 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10416597

RESUMO

The family of vascular endothelial growth factor (VEGF) proteins include potent and specific mitogens for vascular endothelial cells that function in the lation of angiogenesis Inhibition of VEGF-induced angiogenesis either by neutralizing antibodies or dominant-negative soluble receptor, blocks the growth of primary and metastatic experimental tumors Here we report that VEGF expression is induced in Lewis lung carcinomas (LLCs) both in vitro and vivo after exposure to ionizing radiation (IR) and in human tumor cell lines (Seg-1 esophageal adenocarcinoma, SQ20B squamous cell carcinoma, T98 and U87 glioblastomas, and U1 melanoma) in vitro. The biological significance of IR-induced VEGF production is supported by our finding that treatment of tumor-bearing mice (LLC, Seg-1, SQ20B, and U87) with a neutralizing antibody to VEGF-165 before irradiation is associated with a greater than additive antitumor effect. In vitro, the addition of VEGF decreases IR-induced killing of human umbilical vein endothelial cells, and the anti-VEGF treatment potentiates IR-induced lethality of human umbilical vein endothelial cells. Neither recombinant VEGF protein nor neutralizing antibody to VEGF affects the radiosensitivity of tumor cells These findings support a model in which induction of VEGF by IR contributes to the protection of tumor blood vessels from radiation-mediated cytotoxicity and thereby to tumor radioresistance.


Assuntos
Anticorpos Monoclonais/farmacologia , Fatores de Crescimento Endotelial/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linfocinas/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias Experimentais/radioterapia , Neovascularização Patológica/fisiopatologia , Radiossensibilizantes/farmacologia , Estresse Fisiológico/fisiopatologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma/radioterapia , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/radioterapia , Células Cultivadas , Meios de Cultivo Condicionados , Fatores de Crescimento Endotelial/imunologia , Fatores de Crescimento Endotelial/fisiologia , Endotélio Vascular/citologia , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/radioterapia , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Glioblastoma/radioterapia , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Linfocinas/imunologia , Linfocinas/fisiologia , Melanoma/genética , Melanoma/patologia , Melanoma/radioterapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Proteínas de Neoplasias/imunologia , Proteínas de Neoplasias/fisiologia , Transplante de Neoplasias , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/complicações , Neoplasias Experimentais/fisiopatologia , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Tolerância a Radiação/efeitos dos fármacos , Estresse Fisiológico/genética , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/efeitos da radiação , Ensaio Tumoral de Célula-Tronco , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
10.
Cancer Res ; 60(24): 6958-63, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11156396

RESUMO

We examined the effects of a new antiangiogenic isocoumarin, NM-3, as a radiation modifier in vitro and in vivo. The present studies demonstrate that NM-3 is cytotoxic to human umbilical vein endothelial cells (HUVECs) but not to Lewis lung carcinoma (LLC) cells nor Seg-1, esophageal adenocarcinoma cells, in clonogenic survival assays. When HUVEC cultures are treated with NM-3 combined with ionizing radiation (IR), additive cytotoxicity is observed. In addition, the combination of NM-3 and IR inhibits HUVEC migration to a greater extent than either treatment alone. The effects of treatment with NM-3 and IR were also evaluated in tumor model systems. C57BL/6 female mice bearing LLC tumors were given injections for 4 consecutive days with NM-3 (25 mg/kg/day) and treated with IR (20 Gy) for 2 consecutive days. Combined treatment with NM-3 and IR significantly reduced mean tumor volume compared with either treatment alone. An increase in local tumor control was also observed in LLC tumors in mice receiving NM-3/IR therapy. When athymic nude mice bearing Seg-1 tumor xenografts were treated with NM-3 (100 mg/kg/day for 4 days) and 20 Gy (four 5 Gy fractions), significant tumor regression was observed after combined treatment (NM-3 and IR) compared with IR alone. Importantly, no increase in systemic or local tissue toxicity was observed after combined treatment (NM-3 and IR) when compared with IR alone. The bioavailability and nontoxic profile of NM-3 suggests that the efficacy of this agent should be tested in clinical radiotherapy.


Assuntos
Cumarínicos/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Adenocarcinoma/tratamento farmacológico , Animais , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Movimento Celular/efeitos dos fármacos , Movimento Celular/efeitos da radiação , Células Cultivadas , Colágeno/metabolismo , Cumarínicos/toxicidade , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Combinação de Medicamentos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/efeitos da radiação , Neoplasias Esofágicas/tratamento farmacológico , Feminino , Humanos , Isocumarinas , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Transplante de Neoplasias , Proteoglicanas/metabolismo , Radiação Ionizante , Fatores de Tempo , Células Tumorais Cultivadas , Veias Umbilicais/citologia , Veias Umbilicais/efeitos dos fármacos , Veias Umbilicais/efeitos da radiação
11.
Clin Cancer Res ; 6(2): 737-42, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10690561

RESUMO

Although clonogenic or divisional death is the main mechanism by which DNA-damaging agents demonstrate antitumor activity, recent data indicate that strategies specifically designed to trigger apoptosis may also prove to be useful antitumor agents. Protein kinase C (PKC) isoenzymes are involved in the regulation of cell proliferation, differentiation, and survival. Whereas pharmacological inhibition of PKC activity triggers apoptosis in most mammalian cells, cell line and tissue differences in sensitivities to these inhibitors remain. Whereas PKC inhibitors have potential as antitumor agents, issue of kinase specificity and solubility have remained obstacles to their clinical use. In this report, we investigated the antitumor activity of the PKC inhibitor chelerythrine chloride (chelerythrine), a selective inhibitor of group A and B PKC isoforms. Chelerythrine exhibited cytotoxic activity against nine human tumor cell lines tested in vitro. On the basis of the finding that radioresistant and chemoresistant squamous cell carcinoma lines (HNSCC) undergo apoptosis rapidly after treatment with chelerythrine in vitro, we assessed the effects of this agent on p53-deficient SQ-20B HNSCC cells in vivo. The results demonstrate that chelerythrine treatment of nude mice bearing SQ-20B is associated with significant tumor growth delay. Significantly, treatment with chelerythrine resulted in minimal toxicity. These findings demonstrate a potential for chelerythrine as an antitumor drug against squamous cell carcinoma.


Assuntos
Antineoplásicos/toxicidade , Carcinoma de Células Escamosas/patologia , Neoplasias de Cabeça e Pescoço/patologia , Fenantridinas/toxicidade , Alcaloides , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Benzofenantridinas , Peso Corporal/efeitos dos fármacos , Carcinoma de Células Escamosas/tratamento farmacológico , Divisão Celular/efeitos dos fármacos , Inibidores Enzimáticos/toxicidade , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Humanos , Camundongos , Camundongos Nus , Fenantridinas/uso terapêutico , Proteína Quinase C/antagonistas & inibidores , Transplante Heterólogo , Células Tumorais Cultivadas
12.
Cancer Gene Ther ; 5(6): 344-9, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9917088

RESUMO

The purpose of the present study was to determine the therapeutic potential of combining radiotherapy with tumor necrosis factor (TNF)-alpha-based gene therapy in the human prostate cancer PC-3 xenograft. PC-3 cells are highly resistant to TNF-alpha-induced cytotoxicity in vitro. A modest enhancement of radiation killing was observed with the addition of TNF-alpha in clonogenic survival assays. Combined treatment with Ad.Egr-TNF, a replication-deficient adenovirus modified to express TNF-alpha following the exposure of infected cells to ionizing radiation (40 Gy administered at 5 Gy per fraction) in vivo, resulted in increased tumor control, as defined by a reduction of tumor volume, when compared with treatment with Ad.Egr-TNF alone or with radiation alone (P < .03). The improvement in tumor control was achieved without increasing acute normal tissue damage when compared with tissue injury from radiation alone. The results of these studies support further development and clinical application of genetic radiotherapy for human prostate cancer.


Assuntos
Terapia Genética , Neoplasias da Próstata/terapia , Fator de Necrose Tumoral alfa/genética , Adenoviridae/genética , Animais , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Camundongos , Transplante de Neoplasias , Neoplasias da Próstata/radioterapia , Transfecção , Transplante Heterólogo , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/uso terapêutico
13.
Int J Oncol ; 19(4): 833-5, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11562763

RESUMO

We examined the interaction between Alimta and ionizing radiation (IR) as a potential strategy to enhance the therapeutic ratio of combined-modality cancer treatment. Mice bearing human esophageal adenocarcinoma xenografts (Seg-1) or squamous cell carcinoma xenografts (SQ-20B) were treated with Alimta and IR employing a fractionated treatment schedule. Treatment with Alimta alone slowed the growth of Seg-1 but not SQ-20B tumors compared with control tumors. In Seg-1 xenografts combined treatment with Alimta and IR produced significant tumor growth inhibition compared with Alimta alone or IR alone. In SQ-20B xenografts, treatment with Alimta did not enhance IR-mediated tumor growth inhibition suggesting that sensitivity to Alimta is necessary for an interactive cytotoxic effect with IR. The present data suggest the potential clinical efficacy of combining Alimta administration with radiotherapy for Alimta-sensitive cells and indicate that further testing needs to be conducted to optimize the dosing schedule to enhance the interaction between the therapeutic agents.


Assuntos
Antineoplásicos/uso terapêutico , Antagonistas do Ácido Fólico/uso terapêutico , Glutamatos/uso terapêutico , Guanina/análogos & derivados , Guanina/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Adenocarcinoma/radioterapia , Animais , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/radioterapia , Terapia Combinada , Relação Dose-Resposta à Radiação , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/radioterapia , Feminino , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/radioterapia , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/patologia , Pemetrexede , Radiação Ionizante , Transplante Heterólogo , Células Tumorais Cultivadas
14.
Life Sci ; 59(1): 51-60, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8684271

RESUMO

The effect of ethanol (ETOH) exposure on the IGF type II receptor concentration was examined in 18 and 20 day fetal rat tissues. Pregnant dams were fed an ETOH (36% of calories derived from ETOH; 6.6% v/v) liquid diet. Control fetuses were offspring of dams either pair-fed a control liquid diet or ad libitum-fed a standard pelleted lab chow. Fetal brain, heart, kidney, liver, lung and skeletal muscle were removed and whole homogenates from individual animals were analyzed. Results of immunoquantification of IGF type II receptors in whole tissue homogenates show that there is a trend towards increased receptor concentration between 18 and 20 days in all tissue and this trend is significant for lung, liver and muscle. There were no significant differences in receptor concentration between treatment groups. These studies suggest that during the later stages of fetal development, there is an increase in total IGF type II receptors and this increase is undisturbed by ETOH exposure.


Assuntos
Etanol/farmacologia , Feto/efeitos dos fármacos , Receptor IGF Tipo 2/metabolismo , Animais , Feminino , Gravidez , Ratos , Ratos Sprague-Dawley , Receptor IGF Tipo 2/efeitos dos fármacos
15.
Alcohol ; 8(1): 7-11, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-2006988

RESUMO

The influence of acute ethanol (ETOH) on the regulation of GH release has not been clearly elucidated. In this study the effect of ETOH on clonidine (CLON)-induced GH secretion was examined. To test the effect of ETOH on CLON-induced GH release doses of 1, 2, and 3 g/kg ETOH in a 25% ETOH/saline solution or 10 ml/kg saline were injected IP 24 and 1 h before CLON (30 micrograms/kg BW IV). Blood samples were withdrawn through a chronic jugular cannula. ETOH was found to alter the GH surge, which occurred 15 min after CLON administration in controls, in a dose-dependent manner. The 1 g/kg dose reduced the GH surge slightly but not significantly. The 2 g/kg dose suppressed the GH surge which was significantly lower than in controls. The 3 g/kg dose eliminated the GH surge completely. To determine if pituitary GH release is directly influenced by ETOH, animals were injected with GRH (250 ng/kg IV) one hour after the second dose of ETOH (3 g/kg IP). There was no difference in the GH surge in control or ETOH-injected animals. Anti-SRIF administered 30 minutes before ETOH or saline did not alter the response to GRH. These results suggest that ETOH reduces the GH responsiveness to alpha 2-GRH stimulation.


Assuntos
Clonidina/farmacologia , Etanol/farmacologia , Hormônio do Crescimento/metabolismo , Animais , Relação Dose-Resposta a Droga , Masculino , Hormônios Liberadores de Hormônios Hipofisários/farmacologia , Ratos , Ratos Endogâmicos
16.
Cancer Gene Ther ; 16(4): 373-81, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18974777

RESUMO

TNFerade is a radioinducible adenoviral vector expressing tumor necrosis factor-alpha (TNF-alpha) (Ad.Egr-TNF) currently in a phase III trial for inoperable pancreatic cancer. We studied B16-F1 melanoma tumors in TNF receptor wild-type (C57BL/6) and deficient (TNFR1,2-/- and TNFR1-/-) mice. Ad.Egr-TNF+IR inhibited tumor growth compared with IR in C57BL/6 but not in receptor-deficient mice. Tumors resistant to TNF-alpha were also sensitive to Ad.Egr-TNF+IR in C57BL/6 mice. Ad.Egr-TNF+IR produced an increase in tumor-associated endothelial cell apoptosis not observed in receptor-deficient animals. Also, B16-F1 tumors in mice with germline deletions of TNFR1,2, TNFR1 or TNF-alpha, or in mice receiving anti-TNF-alpha exhibited radiosensitivity. These results show that tumor-associated endothelium is the principal target for Ad.Egr-TNF radiosensitization and implicate TNF-alpha signaling in tumor radiosensitivity.


Assuntos
Terapia Genética/métodos , Melanoma Experimental/terapia , Radiossensibilizantes , Fator de Necrose Tumoral alfa/metabolismo , Terapia por Raios X , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Etanercepte , Humanos , Imunoglobulina G/farmacologia , Imunossupressores/farmacologia , Camundongos , Transplante de Neoplasias , Receptores do Fator de Necrose Tumoral , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo II do Fator de Necrose Tumoral/deficiência , Fator de Necrose Tumoral alfa/antagonistas & inibidores
17.
Cancer Gene Ther ; 15(3): 133-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18157146

RESUMO

We report the anticarcinogenic, anti-aging polyphenol resveratrol activates the radio- and chemo-inducible cancer gene therapy vector Ad.Egr.TNF, a replication-deficient adenovirus that expresses human tumor necrosis factor alpha (TNF-alpha) under control of the Egr-1 promoter. Like ionizing radiation or chemotherapeutic agents previously shown to activate Ad.Egr.TNF, resveratrol also induces Egr-1 expression from its chromosomal locus with a possible role for Egr-1 promoter CC(A+T)richGG sequences in the expression of TNF-alpha. Resveratrol induction of TNF-alpha in Ad.Egr.TNF-infected tumor xenografts demonstrated antitumor response in human and rat tumor models comparable to that of radio- or chemotherapy-induced TNF-alpha. Although sirtuins are known targets of resveratrol, in vitro inhibition of SIRT1 activity did not abrogate resveratrol induction of Egr-1 expression. This suggests that SIRT1 is not essential to mediate resveratrol induction of Egr-1. Nevertheless, control of transgene expression via resveratrol activation of Egr-1 may extend use of Ad.Egr.TNF to patients intolerant of radiation or cytotoxic therapy and offer a novel tool for development of other inducible gene therapies.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Estilbenos/farmacologia , Fator de Necrose Tumoral alfa/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Acetilação/efeitos dos fármacos , Animais , Western Blotting , Linhagem Celular Tumoral , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Ensaio de Imunoadsorção Enzimática , Etoposídeo/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Ratos , Resveratrol , Sirtuínas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo
18.
Alcohol Clin Exp Res ; 18(1): 35-41, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8198224

RESUMO

This study examines the effect of ethanol (ETOH) exposure and nutrient restriction on the release of insulin-like growth factor (IGF)-II from 18- and 20-day explanted fetal organs. Fetuses were exposed to ETOH (E) in utero by feeding dams a 36% (calories derived from ETOH: 6.6% v/v) ETOH liquid diet. Control fetuses were offsprings of dams either pair-fed (P) a control liquid diet or ad libitum (A) fed a standard pelleted lab chow. Brain, heart, kidney, liver, lung, muscle, and placenta of fetuses from the same litter were pooled and explanted, and IGF-II concentration in explanted media was analyzed by radioimmunoassay. Maternal and fetal weights were determined during pregnancy and at sacrifice, respectively, to evaluate the influence of ETOH on growth. Both maternal and fetal weights were substantially reduced by ETOH on 18 and 20 days of gestation compared with both A and P controls. At 18 days of gestation, E fetuses (1.33 +/- 0.03 g) weighed less than either A (1.47 +/- 0.03 g) or P (1.54 +/- 0.04 g) fetuses. By 20 days, A mean fetal weight (4.19 +/- 0.23 g) was significantly greater than both P (3.74 +/- 0.06 g) and E (3.28 +/- 0.06 g) fetuses. IGF-II concentration in media from 18-day fetal explants was highest from E (brain, heart, liver, and placenta) and P tissues (kidney, lung, and muscle). IGF-II in media from A tissues (except placenta) was lower than both E and P levels.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/fisiopatologia , Feto/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Animais , Desenvolvimento Embrionário e Fetal/fisiologia , Feminino , Idade Gestacional , Masculino , Técnicas de Cultura de Órgãos , Gravidez , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
19.
Alcohol Clin Exp Res ; 17(6): 1201-6, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7509575

RESUMO

Maternal ethanol (ETOH) exposure is associated with impaired fetal growth. Because insulin-like growth factors (IGFs) are thought to be important in the regulation of fetal somatic growth, we examined the influence of maternal ETOH exposure on fetal growth and plasma levels of IGF-I, IGF-II, and IGF binding proteins (IGFBPs) in the rat model. Control (A) dams were fed a standard rat chow ad libitum. ETOH (E) consuming dams were fed a 36% ETOH diet, and pair-fed (P) dams were fed isocaloric amounts of a control liquid diet. All animals were killed on day 20 of gestation. Plasma concentrations of IGF-I and -II were determined by radioimmunoassay after formic acid-acetone extraction and heat inactivation of IGFBPs. Levels of IGFBPs in fetal plasma were estimated by Western ligand blotting after protein separation by SDS-PAGE and electrotransfer to nitrocellulose. Membranes were probed with [125I]IGF-I, and IGFBPs were identified by autoradiography, quantified by scanning densitometry and results expressed relative to corresponding IGFBPs in control fetal plasma. Maternal weight gain from conception to 20 days of pregnancy was reduced for E compared to P and A dams (p < 0.05 E vs. P or A). The same pattern was reflected in fetal weight that tended to be lower in P compared with A pups, and was significantly reduced in E pups compared with both groups (p < 0.0001 E vs. P or A). Thus, fetal growth was more retarded in E animals despite equal caloric and protein intake by E and P dams.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Proteínas de Transporte/sangue , Transtornos do Espectro Alcoólico Fetal/sangue , Fator de Crescimento Insulin-Like II/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Etanol/toxicidade , Feminino , Retardo do Crescimento Fetal/sangue , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley
20.
Radiat Oncol Investig ; 5(5): 220-6, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9372544

RESUMO

Intratumoral injection of an adenoviral vector containing radiation-inducible DNA sequences of the early growth response gene (Egr-1) promoter ligated to a cDNA encoding tumor necrosis factor-alpha (TNF-alpha; Ad.Egr-TNF) increases the radiation killing of a human radioresistant xenograft (SQ-20B). Viral dose-escalation experiments demonstrated that SQ-20B growth inhibition correlated with viral titer. Injection of 5 x 10(8) PFU Ad.Egr-TNF produced regression to a mean volume of 22 +/- 13% of the original tumor volume, 1 x 10(8) PFU to a mean of 62 +/- 24%, and 5 x 10(7) PFU to a mean of 67 +/- 27%. No regression was observed when tumors were injected with 1 x 10(7) PFU Ad.Egr-TNF or with the null viral vector (Ad.null). When two injections of vector (2 x 10(8) PFU Ad.Egr-TNF) were combined with 50 Gy, a significant increase in tumor regression was observed compared with injection of buffer, Ad.Egr-TNF, or 50 Gy. The interactive killing between TNF and radiation was enhanced significantly (P = 0.05) when the number of injections was increased from two to five while maintaining a constant viral titer (2 x 10(8) PFU Ad.Egr-TNF) and a constant radiation dose (50 Gy). Significant TNF-alpha levels were present in irradiated vs. unirradiated tumors following injection with Ad.Egr-TNF. Taken together, these data suggest that the volumetric reduction produced by the combined effects of Ad.Egr-TNF and radiation is enhanced with increasing vector concentration and the number of vector injections.


Assuntos
Carcinoma de Células Escamosas/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Proteínas Imediatamente Precoces , Adenoviridae , Animais , Carcinoma de Células Escamosas/radioterapia , Terapia Combinada/métodos , Proteínas de Ligação a DNA/farmacologia , Proteína 1 de Resposta de Crescimento Precoce , Feminino , Humanos , Injeções Intralesionais , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fatores de Transcrição/farmacologia , Transplante Heterólogo , Fator de Necrose Tumoral alfa/farmacologia , Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA