Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Gene Ther ; 19(7): 742-51, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21900961

RESUMO

The safety and toxicokinetics of SCH 721015, an adenovirus encoding the human interferon alpha-2b gene, and Syn3 (SCH 209702), a novel excipient, were assessed in cynomolgus monkeys administered intravesical doses of 2.5 × 10E11 or 1.25 × 10E13 particles SCH 721015 in 25 mg Syn3 or 25 mg Syn3 alone on study days 1 and 91. There was no systemic toxicity. Monkeys dosed with SCH 721015 in Syn3 were positive for SCH 721015-specific DNA in the urine for 2 to 3 days following each dose and had interferon alpha-2b protein in the urine for 1-3 days after a single dose and in fewer animals after a second dose. Intracystic administration was associated with inflammation and focal/multifocal ulceration in the urinary bladder and irritation in the ureters and urethra at necropsy. The physical trauma from catheterization and filling/emptying of the bladder was likely a contributing factor and Syn3 exacerbated the trauma. There was nearly complete resolution of these findings 2 months after the last dose. The trauma to the bladder likely contributed to low, transient systemic exposure to Syn3, SCH 721015 and human interferon protein. The results of this study support the clinical investigation of SCH 721015 in Syn3.


Assuntos
Adenoviridae/genética , Ácidos Cólicos/efeitos adversos , Dissacarídeos/efeitos adversos , Técnicas de Transferência de Genes/efeitos adversos , Interferon-alfa/genética , Adenoviridae/imunologia , Administração Intravesical , Animais , Feminino , Humanos , Interferon alfa-2 , Interferon-alfa/sangue , Interferon-alfa/imunologia , Interferon-alfa/urina , Macaca fascicularis , Masculino , Proteínas Recombinantes/sangue , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/urina , Bexiga Urinária/efeitos dos fármacos
2.
Appl Radiat Isot ; 184: 110190, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35303628

RESUMO

A new proton beam-line dedicated to R&D programs has been developed at CentreAntoine Lacassagne (CAL), in Nice (France), in collaboration with the Centrenational d'études spatiales (CNES). This is the second beam-line of the MEDICYC 65 MeV cyclotron that is currently in operation, the first being the clinical 'eye-line' used for ocular proton therapy. The R&D beam-line is proposed with two configurations, the first producing a Gaussian narrow beam of a few mm width, the second a 100 mm diameter flat beam with a homogeneity better than ±3%. The energy range is (20 - ∼60) MeV, where the exact upper limit depends on the beam configuration being used. The energy spread of the non-degraded beam is (0.3 ± 0.1) MeV. A beam current between 10 pA and 10 µA can be produced with a stability better than 0.2% above 100 pA, and 2% below. The beam can be monitored online at a precision better than 5% in the flux range 1E5 (1E6) - 1E9 (1E10) p/cm2/s for a flat (Gaussian) configuration, although work is in progress to extend this range. Targeted applications for the R&D beam-line are instrumentation research, radiation tolerance tests of components and radiobiology.


Assuntos
Terapia com Prótons , Prótons , Ciclotrons , Terapia com Prótons/métodos , Radiobiologia , Dosagem Radioterapêutica , Pesquisa
3.
Cytokine Growth Factor Rev ; 18(5-6): 389-94, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17692556

RESUMO

Intravesical administration of interferon alpha-2b protein (IFN) has been successfully used in the treatment of patients with superficial bladder tumors. Local dosing of IFN minimizes well-known systemic side effects of the drug, but exposure to bladder tumors is limited by the duration of instillation and transient concentrations achieved in the urothelium. Intravesical delivery of the gene encoding interferon results in an alternative strategy for IFN-based therapy of the disease, enabling sustained exposure of IFN protein that results from production by tumor and non-tumor cells in the urothelium. Efficient gene delivery and expression of IFN has been achieved using a recombinant adenovirus gene delivery system (rAd-IFN) in conjunction with the novel small molecule excipient Syn3. Studies with rAd-IFN/Syn3 in animal models result in urine concentrations of IFN that persisted for weeks and correlated with potent anti-tumor effects. The objective of this review is to communicate the rationale and preclinical findings that support ongoing clinical investigation of intravesical rAd-IFN/Syn3 in superficial bladder cancer.


Assuntos
Ácidos Cólicos/administração & dosagem , Dissacarídeos/administração & dosagem , Excipientes/administração & dosagem , Interferon-alfa/administração & dosagem , Interferon-alfa/genética , Neoplasias da Bexiga Urinária/terapia , Adenoviridae/genética , Administração Intravesical , Animais , Ácidos Cólicos/uso terapêutico , Dissacarídeos/uso terapêutico , Excipientes/uso terapêutico , Terapia Genética , Humanos , Interferon alfa-2 , Interferon-alfa/uso terapêutico , Proteínas Recombinantes , Neoplasias da Bexiga Urinária/metabolismo , Urotélio/metabolismo
4.
Handb Exp Pharmacol ; (181): 183-219, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18071947

RESUMO

The biology of the human epidermal growth factor (EGF) receptor-2 (HER2) has been reviewed numerous times and provides an excellent example for developing a targeted cancer therapeutic. Herceptin, the FDA-approved therapeutic monoclonal antibody against HER2, has been used to treat over 150,000 women with breast cancer. However, the developmental history of Herceptin, the key events within the program that created pivotal decision points, and the reasons why decisions were made to pursue the monoclonal antibody approach have never been adequately described. The history of Herceptin is reviewed in a way which allows the experience to be shared for the purposes of understanding the drug discovery and development process. It is the objective of this review to describe the pivotal events and explain why critical decisions were made that resulted in the first therapeutic to successfully target tyrosine kinases in cancer. New approaches and future prospects for therapeutics targeting the HER family are also discussed.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Imunoterapia/métodos , Receptor ErbB-2/imunologia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/história , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Antineoplásicos/química , Antineoplásicos/história , Antineoplásicos/farmacologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Desenho de Fármacos , Fator de Crescimento Epidérmico/metabolismo , Feminino , História do Século XX , História do Século XXI , Humanos , Imunoterapia/história , Modelos Moleculares , Conformação Proteica , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trastuzumab , Resultado do Tratamento
5.
J Clin Invest ; 95(6): 2853-61, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7769126

RESUMO

Culture filtrates of Shigella flexneri 2a strain M4243 grown in iron-depleted medium, caused significant fluid accumulation in rabbit ileal loops. Also, when tested in Ussing chambers, a greater rise in potential difference and short circuit current was seen with such filtrates compared with the medium control. Analogous filtrates from two M4243 derivatives lacking the 140-MD invasiveness plasmid (either M4243avir or BS103) retained 60-65% of the wild-type enterotoxic activity. Ultrafiltration and gel exclusion size fractionation of M4243 filtrate revealed that the activity was approximately 60 kD. SDS-PAGE performed on this fraction showed 18 bands, 5 of which reacted with human convalescent sera. Genes encoding this enterotoxin, named ShET1 for Shigella enterotoxin 1, were cloned from the S. flexneri 2a chromosome, and two separate open reading frames of 534 and 186 bp were sequenced. These observations suggest that S. flexneri 2a elaborates two distinct enterotoxins: ShET1, encoded by genes located on the chromosome, and ShET2, encoded by a gene on the 140-MD invasiveness plasmid. ShET1, which is composed of two distinct subunits and is elaborated in vivo, where it elicits an immune response, may be important in the pathogenesis of diarrheal illness due to S. flexneri 2a.


Assuntos
Enterotoxinas/genética , Shigella flexneri/patogenicidade , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Enterotoxinas/farmacologia , Genes Bacterianos , Intestino Delgado/efeitos dos fármacos , Masculino , Dados de Sequência Molecular , Coelhos , Shigella flexneri/genética
6.
Cancer Gene Ther ; 13(7): 664-75, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16514422

RESUMO

Type I interferons (e.g. IFNalpha2b) have been successfully used to treat a variety of hematological malignancies, but have not been efficacious for treatment of most solid tumors. We tested the hypothesis that delivery of type I interferon utilizing recombinant adenovirus (rAd) vectors may improve treatment efficacy of metastatic carcinomas by providing increased interferon exposure resulting from continuous transgene expression. Treatment of mice with a rAd-vector expressing hybrid-IFN (rAd-IFNalpha2alpha1) inhibited 4T1 mammary carcinoma tumor growth and induced tumor regression in a dose-dependent manner. Moreover, rAd-IFNalpha2alpha1 treatment reduced hepatic and pulmonary metastatic burden. A comparison of local and systemic routes of administration demonstrated that intratumoral delivery of rAd-IFNalpha2alpha1 was sufficient for inhibition of tumor growth. Moreover, it reduced toxicity associated with high-dose systemic IFNalpha2alpha1 exposure. Interestingly, antitumor activity following intratumoral treatment was due, in part, to the immunostimulatory capacity of the rAd vector component. Furthermore, systemic administration of rAd-IFNalpha2alpha1 potentiated the immunotherapeutic effect induced by local intralesional delivery of empty-rAd vector. These results suggest continuous interferon-alpha exposure may provide improved antitumor responses for metastatic carcinomas. Additionally, immunostimulatory responses induced by rAd-IFNalpha2alpha1 may mitigate the immune-evasive tumor microenvironment.


Assuntos
Adenoviridae , Carcinoma/terapia , Terapia Genética , Vetores Genéticos , Interferon-alfa , Animais , Carcinoma/genética , Carcinoma/imunologia , Carcinoma/patologia , Relação Dose-Resposta Imunológica , Feminino , Imunoterapia , Interferon alfa-2 , Interferon-alfa/genética , Interferon-alfa/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes
7.
Cancer Gene Ther ; 13(2): 169-81, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16082381

RESUMO

The major focus of intrahepatic arterial (IHA) administration of adenoviruses (Ad) has been on safety. Currently, there is little published data on the biological responses to Ad when administered via this route. As part of a Phase I study, we evaluated biological responses to a replication-defective adenovirus encoding the p53 transgene (SCH 58500) when administered by hepatic arterial infusion to patients with primarily colorectal cancer metastatic to the liver. In analyzing biological responses to the Ad vector, we found that both total and neutralizing Ad antibodies increased weeks after SCH 58500 infusion. The fold increase in antibody titers was not dependent on SCH 58500 dosage. The proinflammatory cytokine interleukin-6 (IL-6) transiently peaked within 6 h of dosing. The cytokine sTNF-R2 showed elevation by 24 h post-treatment, and fold increases were directly related to SCH 58500 doses. Cytokines TNF-alpha, IL-1beta, and sTNF-R1 showed no increased levels over 24 h. Predose antibody levels did not appear to predict transduction, nor did serum Ad neutralizing factor (SNF). Delivery of SCH 58500 to tumor tissue occurred, though we found distribution more predominantly in liver tissues, as opposed to tumors. RT-PCR showed significantly higher expression levels (P<0.0001, ANOVA) for adenovirus type 2 and 5 receptor (CAR) in liver tissues, suggesting a correlation with transduction. Evidence of tumor-specific apoptotic activity was provided by laser scanning cytometry, which determined a coincidence of elevated nuclear p53 protein expression with apoptosis in patient tissue. IHA administration of a replication defective adenovirus is a feasible mode of delivery, allowing for exogenous transfer of the p53 gene into target tissues, with evidence of functional p53. Limited and transient inflammatory responses to the drug occurred, but pre-existing immunity to Ad did not preclude SCH 58500 delivery.


Assuntos
Adenoviridae/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Genes p53/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Neoplasias Hepáticas/secundário , Adulto , Idoso , Análise de Variância , Anticorpos Antivirais/sangue , Apoptose/efeitos dos fármacos , Neoplasias Colorretais/imunologia , Citocinas/sangue , Primers do DNA , Feminino , Artéria Hepática , Humanos , Infusões Intra-Arteriais , Citometria de Varredura a Laser , Masculino , Pessoa de Meia-Idade , Receptores Virais/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Cancer Res ; 52(7): 1916-23, 1992 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-1348016

RESUMO

The HER-2/neu oncogene encodes a Mr 185,000 transmembrane phosphoglycoprotein which is overexpressed in 25-35% of breast and ovarian neoplasms and portends a poor prognosis. We have studied the feasibility of targeting this oncoprotein, designated p185, with radioiodinated murine monoclonal antibodies (muMABs) 4D5 and 7C2, which recognize distinct epitopes on its extracellular domain. The rates of internalization and catabolism of these antibodies were analyzed by cellular radioimmunoassay and electron microscopy. After binding to NIH3T3 HER-2/neu cells, which show high surface expression of p185, the muMABs were endocytosed via coated pits, routed to lysosomes, and degraded. Approximately 44% of 125I-4D5 and 39% of 125I-7C2 were catabolized by tumor cells after 24 h. The biodistribution of radiolabeled 4D5 and 7C2 were evaluated in beige/nude mice bearing s.c. NIH3T3 HER-2/neu grafts. A high specificity of localization was seen with tumor:organ ratios of activity generally ranging from 5:1 to 30:1. However, the percentage injected dose of radioactivity per gram of tumor declined sharply from 25% at 24 h to 5% at 120 h postinjection. Treating the animals with 400-700 muCi 131I-4D5 caused a marked inhibition of tumor growth, although no mice were cured. Unlabeled 4D5 had no effect on tumor progression in this model, but administering 400-700 muCi of 131I-DA4-4, an isotype-matched irrelevant muMAB, resulted in an intermediate degree of growth retardation. Analysis of kinetic blood data and whole-body time-activity curves indicated that the irrelevant conjugate remained in the body 2-3 times longer than 131I-4D5. Radioiodinated anti-HER-2/neu muMABs are attractive agents for radioimmunodiagnosis and radioimmunotherapy of aggressive HER-2/neu-positive breast and ovarian carcinomas, but effective strategies for retarding intratumoral catabolism may be necessary to optimize their clinical utility.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Transformação Celular Neoplásica , Radioisótopos do Iodo/uso terapêutico , Proteínas Tirosina Quinases/imunologia , Proteínas Proto-Oncogênicas/imunologia , Proto-Oncogenes , Células 3T3 , Animais , Autorradiografia , Neoplasias da Mama , Feminino , Imunoglobulina G , Masculino , Camundongos , Camundongos Nus , Microscopia Imunoeletrônica , Transplante de Neoplasias , Neoplasias Ovarianas , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/genética , Radioimunoterapia , Receptor ErbB-2 , Distribuição Tecidual , Transfecção
9.
Cancer Res ; 50(12): 3493-502, 1990 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-2140289

RESUMO

[6RS]Leucovorin (5-formyltetrahydrofolate; 5-CHO-H4PteGlu) administered in different regimens in combination with 5-fluorouracil (FUra) has increased the response rates to FUra in patients with colon adenocarcinoma. Using preclinical models of human colon adenocarcinomas as xenografts in immune-deprived mice, the effect of the rate of administration of racemic [6RS]leucovorin on the concentration-time profile of reduced folates in plasma, size of intratumor pools of 5,10-methylenetetrahydrofolates (CH2-H4PteGlun) and tetrahydrofolates (H4PteGlun), and the distribution of their polyglutamate species have been examined. Bolus injection i.v., or 4-h or 24-h infusion of [6RS]leucovorin (500 mg/m2) yielded similar concentration profiles of the biologically active [6S] and inactive [6R] isomers of 5-CHO-H4-PteGlu and 5-methyltetrahydrofolate (5-CH3-H4PteGlu) in mouse plasma to those previously reported in humans, but with more rapid elimination half-lives (t1/2 = 11 to 16 min, 23 to 41 min, and 30 to 35 min, respectively). Thus, reduced folates remained elevated in plasma during the period of [6RS]leucovorin administration. In HxELC2 and HxGC3 tumors, pools of CH2-H4PteGlun and H4PteGlun were increased from 350% to 700% of control, but only during [6RS]leucovorin infusion. Intracellular levels subsequently declined rapidly, similar to the loss of reduced folates from plasma. Increasing the rate of [6RS]leucovorin delivery by decreasing the time for administration from a 24-h to a 4-h infusion did not further increase the intratumor pools of CH2-H4PteGlun and H4PteGlun, suggesting saturation in the cellular metabolism of [6RS]leucovorin. In HxGC3 tumors, CH2-H4PteGlu4-5 were elevated more rapidly than in line HxELC2, which accumulated predominantly a shorter chain length species following i.v. bolus injection. During the 4-h infusion schedule, di- and triglutamate species in particular accumulated in both tumors with no elevation in CH2-H4PteGlu5 until the infusion was discontinued, when this species increased as the shorter chain length forms were declining. However, during the 24-h infusion of [6RS]leucovorin, CH2-H4PteGlu3-5 were elevated in tumors. Since these species have been reported to increase the binding affinity of [6-3H]5-fluorodeoxyuridine monophosphate ([6-3H]FdUMP) to thymidylate synthase, and intratumor pools of CH2-H4PteGlun and H4PteGlun were elevated during the 24-h infusion of [6RS]leucovorin, this was considered to be the preferred schedule for administration.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Adenocarcinoma/metabolismo , Neoplasias do Colo/metabolismo , Floxuridina/metabolismo , Leucovorina/farmacologia , Tetra-Hidrofolatos/sangue , Timidilato Sintase/antagonistas & inibidores , Adenocarcinoma/sangue , Animais , Neoplasias do Colo/sangue , Feminino , Floxuridina/administração & dosagem , Floxuridina/sangue , Floxuridina/farmacologia , Meia-Vida , Humanos , Injeções Intravenosas , Leucovorina/administração & dosagem , Leucovorina/sangue , Camundongos , Camundongos Endogâmicos CBA , Timidilato Sintase/sangue , Fatores de Tempo
10.
Oncogene ; 12(8): 1617-23, 1996 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-8622881

RESUMO

Mutations of p53 gene are reported in 50-60% of human cancers and reintroduction of wild-type p53 can suppress cell proliferation. In this study, replication deficient recombinant adenovirus encoding wild-type p53 (ACN53) under the control of the human cytomegalovirus (CMV) promoter was constructed. A specific incorporation of the p53 gene with ACN53 reduced 3 (deleted p53 gene) cells was observed. ACN53 reduced the colony-forming ability of SK-OV-3 cells 72-216 h after single infection. A highly aggressive ovarian xenograft model was established in which animals die between 25-45 days. A localization study with the adenovirus-containing beta galactosidase reporter gene showed effective gene transfer in the tumor tissues. Ex vivo treatment of SK-OV-3 cells with ACN53 followed by injection into nude mice, increased the survival of the p53 treated mice by more than 50% compared with control animals. Gene therapy with ACN53 in intraperitoneal model of SK-OV-3 cells in two independent experiments revealed that there were some long-term survivors in the group of mice [2/5 (66 and 120 days) and [2/8 (166 and 423 days)] treated with ACN53. These findings demonstrate the potential of the p53 tumor suppressor gene therapy in human ovarian carcinoma.


Assuntos
Adenocarcinoma/terapia , Adenoviridae/genética , Genes p53 , Terapia Genética , Neoplasias Ovarianas/terapia , Adenocarcinoma/genética , Adenocarcinoma/virologia , Animais , Testes de Carcinogenicidade , Feminino , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/virologia , Transdução Genética , Células Tumorais Cultivadas
11.
Oncogene ; 10(4): 697-704, 1995 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-7862447

RESUMO

Reconstitution of retinoblastoma gene (RB) deficient tumor cells with RB generally leads to growth suppression in vitro and/or reduced tumorigenicity in nude mice. An alternate approach to gene replacement is the delivery of the RB gene product (p110RB) into cells lacking its expression. In this report we demonstrate that exogenously added p110RB is taken up by and localized to the nucleus of cultured cells and has growth suppression properties similar to endogenous RB. RB-negative (RBneg) tumor cells are preferentially growth inhibited while most RB-positive (RBpos) tumor cells and normal cells are much less sensitive. We have extended these studies to relevant nude mouse xenograft models for human lung cancer. Local or systemic administration of p110RB inhibits tumor growth in treated animals. These results represent the first use of a tumor suppressor protein as a potential cancer therapeutic.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Divisão Celular/efeitos dos fármacos , Neoplasias Experimentais/patologia , Proteína do Retinoblastoma/farmacologia , Animais , Núcleo Celular/metabolismo , Inibidores do Crescimento , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transplante Heterólogo , Células Tumorais Cultivadas
12.
Oncogene ; 17(17): 2235-49, 1998 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-9811454

RESUMO

HER-2 oncogene encodes a transmembrane growth factor receptor that is overexpressed in 25-30% of patients with primary breast and ovarian cancer. A murine monoclonal antibody, 4D5, to the extracellular domain of HER-2 receptor elicits cytostatic growth inhibition of tumor cells overexpressing HER-2 protein, but clinical use of this antibody is limited by genesis of human anti-mouse antibodies. To avoid this problem, a recombinant humanized 4D5 monoclonal antibody (rhuMAb HER-2) was developed and tested using a human tumor xenograft model. Human breast and ovarian cancer cells which overexpress HER-2 were inhibited in vivo by the rhuMAb HER-2 antibody. Tumor growth relative to control was reduced at all doses of antibody tested, and the magnitude of growth inhibition was directly related to dose of rhuMAb HER-2. Tumor growth resumed on termination of antibody therapy, indicating a cytostatic effect. To elicit a cytotoxic response, human breast tumor xenografts were treated with a combination of antibody and antitumor drugs, cisplatin or doxorubicin. The combination of antibody with either cisplatin or doxorubicin resulted in significantly greater growth inhibition, with the cisplatin combination demonstrating a greater response. In addition, therapy with cisplatin and antireceptor antibody elicited complete tumor remissions after 2-3 cycles of therapy. The schedule of administration of anti-receptor antibody and cisplatin was critical for occurrence of antibody-induced potentiation in cisplatin cytotoxicity. Enhanced killing of tumor cells was found only if antibody and drug were given in close temporal proximity. Since interference with DNA repair pathways may contribute to this receptor-enhanced chemosensitivity, repair of cisplatin-damaged reporter DNA (pCMV-beta) was determined in human breast cells. As in studies of antibody-enhanced cisplatin cytotoxicity in vivo, treatment with rhuMAb HER-2 blocked the repair of cisplatin-damaged DNA only if the antibody was administered in close temporal proximity to transfection of the drug-exposed reporter DNA. An alternative measure of DNA repair, unscheduled DNA synthesis, was also assessed. Treatment with either cisplatin or doxorubicin led to an increase in unscheduled DNA synthesis that was reduced by combined therapy with antireceptor antibody specific to HER-2-overexpressing breast cancer cells. Using a direct measure of DNA repair, therapy of HER-2-overexpressing cells with rhuMAb HER-2 also blocked the removal of cisplatin-induced DNA adducts. Expression of p21/WAF1, an important mediator of DNA repair, was disrupted in breast cancer cells with HER-2 overexpression, but not in control cells, after treatment with HER-2 antibody, thus suggesting cross-communication between the HER-2 signaling and DNA repair pathways. These data demonstrate an in vivo antiproliferative effect of rhuMAb HER-2 on tumors that overexpress HER-2 receptor and a therapeutic advantage in the administration of the antireceptor antibody in combination with chemotherapeutic agents.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Mama/terapia , Cisplatino/uso terapêutico , Proteínas de Neoplasias/imunologia , Neoplasias Ovarianas/terapia , Receptor ErbB-2/imunologia , Animais , Neoplasias da Mama/metabolismo , Cisplatino/metabolismo , Terapia Combinada , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Adutos de DNA/metabolismo , Reparo do DNA , DNA de Neoplasias/biossíntese , Doxorrubicina/uso terapêutico , Feminino , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transfecção , Transplante Heterólogo , Células Tumorais Cultivadas
13.
Oncogene ; 15(3): 337-45, 1997 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-9233768

RESUMO

This study analyses whether the inability of p53 to induce G1 arrest after the restriction point relates to an inability to modulate pRb phosphorylation. Transient p53 overexpression in normal human diploid fibroblasts and p53-deficient cancer cells led to increased levels of the cyclin-dependent kinase inhibitor p21 cip1/Waf1/Sdi1 and an accumulation of hypophosphorylated pRb in cells growing asynchronously and in cells synchronized in late G1 or M. Similarly, gamma-irradiation of asynchronous, late-G1, or S phase fibroblasts led to an increase in hypophosphorylated pRb. Experiments with fibroblasts expressing the HPV16 E6 protein indicated that accumulation of hypophosphorylated pRb required functional p53. Progression into and through S phase was not altered by the presence of hypophosphorylated pRb in late G1, consistent with the failure of p53 to mediate G1 arrest in cells that are past the restriction point. These data indicate that accumulation of hypophosphorylated pRb has significantly different effects on cell cycle progression in early G1 versus late G1 or S phase.


Assuntos
Ciclo Celular/fisiologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Ciclinas/biossíntese , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Carcinoma Pulmonar de Células não Pequenas , Ciclo Celular/efeitos da radiação , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Diploide , Inibidores Enzimáticos/metabolismo , Fibroblastos , Fase G1 , Raios gama , Humanos , Neoplasias Pulmonares , Proteínas Oncogênicas Virais/biossíntese , Papillomaviridae/genética , Fosforilação , Proteínas Recombinantes/biossíntese , Proteínas Repressoras/biossíntese , Fase S , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese
14.
Clin Cancer Res ; 4(7): 1649-59, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9676839

RESUMO

p53 tumor suppressor gene therapy has been proposed for cancers characterized by inactivation of p53 function, and successful therapy will require efficient strategies for gene delivery. To maximize transgene expression in tumors, a clinical strategy has been proposed to treat neoplasms in the liver via hepatic artery administration of a recombinant adenovirus encoding wild-type p53 (rAd-p53). We have developed a syngeneic rat model using a p53mut hepatocellular carcinoma cell line (McA-RH7777) that results in multifocal liver tumor nodules to provide experimental support for this strategy. Treatment of McA-RH7777 cells with rAd-p53 in vitro resulted in efficient transgene expression, growth suppression, and apoptosis. Intrahepatic artery dosing with rAd-p53 or an adenovirus encoding beta-galactosidase (rAd-betagal) increased transgene expression in tumor tissue and decreased systemic exposure when compared with i.v. dosing. Daily hepatic artery dosing of rAd-p53 suppressed tumor growth when compared with untreated rats or animals treated with rAd-betagal. These data demonstrate the potential for arterial gene delivery to tumors using recombinant adenoviruses, and support continued investigation of rAd-p53 gene therapy for liver malignancies.


Assuntos
Adenoviridae , Carcinoma Hepatocelular/terapia , Genes p53 , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Neoplasias Hepáticas Experimentais/terapia , Proteínas de Neoplasias/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Carcinoma Hepatocelular/metabolismo , Vírus Defeituosos , Feminino , Expressão Gênica , Artéria Hepática , Humanos , Infusões Intra-Arteriais , Infusões Intravenosas , Neoplasias Hepáticas Experimentais/metabolismo , Proteínas de Neoplasias/genética , Ratos , Transgenes , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
15.
Hum Gene Ther ; 5(9): 1079-88, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7833367

RESUMO

We have constructed recombinant human adenoviruses that express wild-type human p53 under the control of either the Ad 2 major late promoter (MLP) or the human cytomegalovirus (CMV) immediate early gene promoter. Each construct replaces the Ad 5 E1a and E1b coding sequences necessary for viral replication with the p53 cDNA and MLP or CMV promoter. These p53/Ad recombinants are able to express p53 protein in a dose-dependent manner in infected human cancer cells. Tumor suppressor activity of the expressed p53 protein was assayed by several methods. [3H]Thymidine incorporation assays showed that the recombinant adenoviruses were capable of inhibiting DNA synthesis in a p53-specific, dose-dependent fashion. Ex vivo treatment of Saos-2 tumor cells, followed by injection of the treated cells into nude mice, led to complete tumor suppression using the MLP/p53 recombinant. Following a single injection of CMV/p53 recombinant adenovirus into the peritumoral space surrounding an in vivo established tumor derived from a human small cell lung carcinoma cell line (NIH-H69), we were able to detect p53 mRNA in the tumors at 2 and 7 days post-injection. Continued treatment of established H69 tumors with MLP/p53 recombinant led to reduced tumor growth and increased survival time compared to control treated animals. These results indicate that recombinant adenoviruses expressing wild-type p53 may be useful vectors for gene therapy of human cancer.


Assuntos
Adenovírus Humanos/genética , Carcinoma de Células Pequenas/terapia , Vírus Defeituosos/genética , Genes p53 , Terapia Genética , Vetores Genéticos , Neoplasias/terapia , Animais , Sequência de Bases , Carcinoma de Células Pequenas/patologia , Citomegalovirus/genética , Replicação do DNA , DNA Complementar/genética , DNA Recombinante/genética , DNA Viral/genética , Feminino , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Dados de Sequência Molecular , Transplante de Neoplasias , Neoplasias/genética , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão , Transplante Heterólogo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
16.
Cancer Gene Ther ; 5(1): 52-63, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9476967

RESUMO

The last two decades have led to a greater understanding of the genetic basis of human malignancy. Although numerous genetic alterations have been detected in cancer, activation of oncogenes and inactivation of cell cycle regulators (e.g., tumor suppressor genes) are now known to play a critical role in the progression of the disease. Therapeutic strategies based on specific molecular alterations in cancer include reintroduction of wild-type tumor suppressor function to cells lacking the gene. p53 gene therapy provides an attractive strategy to test the potential clinical feasibility of this approach. Alterations in p53 function are present in approximately half of all malignancies, and expression of wild-type p53 can result in apoptosis in human tumor cells. This review summarizes current investigations with p53 gene therapy, highlighting the preclinical efforts with adenoviral, retroviral, and lipid-based gene delivery systems. A comprehensive review of the various clinical targets suggested for p53 gene therapy is presented together with challenges and prospects for future clinical investigation.


Assuntos
Genes p53 , Terapia Genética , Neoplasias/terapia , Terapia Combinada , Vetores Genéticos , Humanos , Neoplasias/genética
17.
Cancer Gene Ther ; 7(2): 197-206, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10770627

RESUMO

Tumor cell contamination of stem cell products can contribute to tumor relapse following high-dose chemotherapy and stem cell rescue. Numerous techniques have been used to remove the tumor cells from stem cell products with the objective of prolonging relapse-free survival. However, to date these techniques have been relatively ineffectual and/or toxic to hematopoietic stem and progenitor cells. The differential infectivity of adenovirus (Adv) vectors for breast cancer cells, compared with hematopoietic cells, has suggested that Adv-p53 might provide an effective purging strategy. To facilitate the use of Adv-p53 as a clinical strategy, we undertook studies to determine the parameters necessary for optimal stem cell product purging. The parameters studied were the particle number to nucleated cell ratio, the duration of coincubation, the incubation volume, and the presence or absence of hematopoietic progenitor cells. We have found that these parameters are interdependent and conclude that a 4-hour coincubation with an Adv-p53 particle to nucleated cell ratio of 2000:1 with 2 x 10(8) nucleated cells/mL is optimal for tumor cell purging. Furthermore, this appeared to be a safe procedure, with total loss of clonogenic growth of breast cancer cells as well as no significant effect on progenitor cell function as determined by granulocyte-macrophage colony-forming unit assays.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/genética , Neoplasias da Mama/virologia , Células-Tronco Hematopoéticas/patologia , Proteína Supressora de Tumor p53/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Morte Celular/genética , Divisão Celular/genética , Separação Celular/métodos , Células Clonais/patologia , Técnicas de Cocultura , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Inibidores do Crescimento/genética , Inibidores do Crescimento/toxicidade , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco , Proteína Supressora de Tumor p53/biossíntese
18.
Cancer Gene Ther ; 4(2): 129-38, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9080122

RESUMO

In response to DNA damage, p53 protein accumulates in the cell nucleus causing cells to undergo DNA repair or apoptosis, programmed cell death. Reintroduction of wild-type p53 into tumors with null or mutant p53 offers a novel strategy for controlling tumor growth, by inducing apoptotic death in neoplastic cells. The efficacy of a replication-deficient p53 adenovirus construct was tested against three human breast cancer cell lines expressing mutant p53, MDA-MB-231, -468, and -435. 231 and 468 cells were both highly transduced at a multiplicity of infection of 10. By contrast, 435 cells were rarely transduced. p53 adenovirus-mediated gene therapy was highly effective against 231 and 468 tumor xenografts in nude mice. At a total dose of 2.2 x 10(9) cellular infectious units (CIU), inhibition of 231 tumor growth was 86% (P < or = .01). Thirty-seven percent of that growth inhibition was due to p53, while 49% was adenovirus-specific. Inhibition of 468 tumor growth was 74% (P < or = .001). Forty-five percent of that inhibition was p53-specific, while 28% was adenovirus-specific. The ED50 values for 231 tumors and 468 tumor growth inhibition were 3 x 10(8) CIU and 2 x 10(8) CIU, respectively. Injection of p53 Ad into 231 or 468 tumors induced apoptosis. By contrast, growth inhibition in 435 tumors treated with p53 adenovirus was not significant, probably due to low adenovirus transduction. 231 and 435 cells both expressed high levels of alpha v, beta 1, beta 3, and beta 5 integrin subunits, ruling out lack of the appropriate integrins as the reason for the low infection rate in 435 cells. Our results demonstrate the ability of wild-type p53 to curtail cancerous cell growth in vivo in tumors expressing mutant p53. The ability of beta-gal Ad to infect tumor cells in vitro was generally predictive of in vivo p53 Ad efficacy.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Genes p53 , Terapia Genética/métodos , Animais , Apoptose/genética , Neoplasias da Mama/patologia , Divisão Celular/genética , Relação Dose-Resposta a Droga , Feminino , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Humanos , Integrinas/biossíntese , Camundongos , Camundongos Nus , Neoplasias Experimentais/genética , Neoplasias Experimentais/terapia , Transdução Genética , Transplante Heterólogo , Células Tumorais Cultivadas
19.
Cancer Gene Ther ; 2(3): 191-7, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8528962

RESUMO

We have investigated whether adenovirally mediated gene transfer of the herpes simplex thymidine kinase gene to human hepatocellular carcinoma (HCC) cell lines can sensitize these cells to the prodrug ganciclovir and thereby provide a therapeutic option for this intractable cancer. Two replication-deficient adenoviruses encoding for the herpes simplex virus type-1 (HSV) thymidine kinase (TK) gene were generated in which expression of TK is under the control of either the human cytomegalovirus immediate early promoter (CMV) or the human alpha-fetoprotein (AFP) promoter/enhancer. We demonstrate that the combination of adenovirally mediated TK gene transfer and ganciclovir treatment effectively inhibits proliferation and causes cell death of HCC cells in vitro and that in vivo TK gene transfer and ganciclovir treatment inhibits hepatocellular tumor growth in a mouse model of this cancer. Furthermore, we show that expression of the TK gene can be restricted to those HCCs that express the tumor marker AFP through the incorporation of the AFP enhancer/promoter within an adenoviral vector.


Assuntos
Carcinoma Hepatocelular/terapia , Resistencia a Medicamentos Antineoplásicos/genética , Terapia Genética , Herpesvirus Humano 1/genética , Timidina Quinase/genética , Animais , Carcinoma Hepatocelular/genética , Divisão Celular/efeitos dos fármacos , Ganciclovir/farmacologia , Vetores Genéticos/farmacologia , Humanos , Camundongos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Timidina Quinase/farmacologia , alfa-Fetoproteínas/biossíntese , alfa-Fetoproteínas/genética
20.
Cancer Gene Ther ; 2(2): 97-104, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7621262

RESUMO

Transitional cell carcinoma (TCC) of the bladder is associated with characterized lesions in dominant and recessive oncogenes. The understanding of the molecular basis of tumorigenesis in these instances makes possible the application of gene therapy strategies for TCC. In this regard, the ability to directly access the epithelium of the genitourinary (GU) tract via the urethra provides a practical means to implement these various gene therapy approaches. We thus explored vector strategies to accomplish direct in vivo transduction of GU epithelium. Initially, three human (HT 1197, HT 1376, T24) and one mouse (MBT-2) TCC cell lines were transduced using a recombinant adenoviral vector expressing the firefly luciferase reporter gene, rAd-CMV-Luc. In these studies, reporter gene expression was found to be significantly elevated above background for all four cell lines. Of note, the TCC cell lines HT 1197 and HT 1376 showed expression levels comparable with the cervical carcinoma cell line HeLa, a cell line previously shown to be highly susceptible to recombinant adenovirus-mediated gene transduction. An in vitro time course for T24 and MBT-2 using rAd-CMV-Luc showed peak expression 1 day after transduction for the T24 line and 3 days after transduction for the MBT-2 line, with detectable levels of expression persisting for at least 7 days. As a next step, human and mouse primary tissue deriving from the GU epithelium were transduced using rAd-CMV-Luc. In this assay, luciferase expression levels significantly above background were observed in both instances.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Adenovírus Humanos/genética , Carcinoma de Células de Transição/terapia , Terapia Genética/métodos , Vetores Genéticos , Proteínas Recombinantes de Fusão/biossíntese , Neoplasias da Bexiga Urinária/terapia , Bexiga Urinária/patologia , Animais , Carcinoma de Células de Transição/genética , Carcinoma de Células de Transição/patologia , Epitélio/metabolismo , Epitélio/virologia , Feminino , Genes Reporter , Humanos , Luciferases/biossíntese , Luciferases/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley , Transfecção , Células Tumorais Cultivadas , Bexiga Urinária/metabolismo , Bexiga Urinária/virologia , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , beta-Galactosidase/biossíntese , beta-Galactosidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA