Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Natl Cancer Inst ; 79(1): 59-65, 1987 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-3298786

RESUMEN

Molecular studies have demonstrated increased expression of the Harvey (Ha) ras oncogene in human breast and colon carcinomas. With the use of a direct-binding liquid competition radioimmunoassay (RIA), capable of providing truly quantitative analysis of the 21,000-dalton (p21) ras oncogene and protooncogene products, absolute levels of Ha-ras p21 have been determined in human breast and colon carcinomas, benign lesions, and/or their respective normal tissues. Enhanced Ha-ras expression was documented in 66% of breast and 100% of colon carcinomas as compared with their normal counterparts, with levels in breast carcinomas ranging from 10.1 to 50.4 pg ras p21/micrograms protein and those in colon carcinomas ranging from 18.4 to 51.7 pg ras p21/micrograms protein. Some dysplastic lesions of the breast and colon also contained elevated Ha-ras p21. Relative levels of Ha-ras p21 expression, detected by competition RIA, correlated with percent Ha-ras p21-positive cells as determined by immunohistochemical assays. By use of liquid competition RIA and immunohistochemical assays, it has been shown that levels of ras p21 expression did not always correlate between primary and metastatic colon lesions of the same patient. The use of the quantitative RIA and semiquantitative immunohistochemical assays, in concert with cDNA probes for identification of specific ras point-mutated oncogenes or protooncogenes, may now provide the means for definitive quantitative analyses of ras p21 in human carcinomas and benign lesions.


Asunto(s)
Adenocarcinoma/genética , Neoplasias de la Mama/genética , Neoplasias del Colon/genética , Oncogenes , Proteínas Proto-Oncogénicas/análisis , Adenocarcinoma/secundario , Neoplasias del Colon/secundario , Histocitoquímica , Humanos , Técnicas Inmunológicas , Proteínas Proto-Oncogénicas p21(ras) , Proto-Oncogenes , Radioinmunoensayo/métodos
2.
J Natl Cancer Inst ; 64(4): 967-75, 1980 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-6154165

RESUMEN

Radioimmunoassays were developed that can detect antigenic determinants common to mammary tumor viruses (MTV's) of four distinct Mus species: M. musculus, M. cervicolor, M. cookll, and M. caroll. The radioimmunoassays were based on the immunologic cross-reactivity observed between the murine mammary tumor viruses (MuMTV) of M. musculus and type B retrovirus isolated from M. cervicolor. Both of the glycoproteins of MuMTV (gp52, gp36) shared antigenic determinants with virions of M. cervicolor mammary tumor virus. Interspecies radioimmunoassays for gp52 and gp36 were developed and used to detect viruses in the milk of noninbred feral Mus species and MuMTV-related translational products in mammary tumors in these species. Type C and type D retroviruses, as well as the M432 retrovirus of M. cervicolor, did not react in either assay. Both interspecies immunoassays were therefore specific for the detection of distinct MuMTV-related antigenic determinants.


Asunto(s)
Antígenos Virales/análisis , Neoplasias Mamarias Experimentales/inmunología , Virus del Tumor Mamario del Ratón/inmunología , Ratones/microbiología , Animales , Reacciones Cruzadas , Epítopos , Glicoproteínas/inmunología , Leche/inmunología , Leche/microbiología , Radioinmunoensayo , Especificidad de la Especie , Proteínas Virales/inmunología
3.
J Natl Cancer Inst ; 61(6): 1509-19, 1978 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-82634

RESUMEN

A virus, similar to the murine mammary tumor viruses (MuMTV) of the laboratory mouse Mus musculus, was identified in the milk of M. cervicolor popaeus mice. The virus was morphologically indistinguishable from the type-B MuMTV and was thus termed MC-MTV. Radioimmunoassays for the 52,000-dalton major envelope glycoprotein and the 28,000-dalton major internal protein of MuMTV demonstrated that MC-MTV shared some antigenic determinants with both of these MuMTV proteins. This reactivity was clearly different, however, from that observed with all MuMTV tested from M. musculus. MC-MTV had a density of 1.16 g/ml in sucrose and a virion-associated DNA polymerase with a divalent cation preference for Mg2+ over Mn2+. Radioimmunoassays clearly differentiated MC-MTV from the other viruses previously identified from M. cervicolor, i.e., M432, CERV-CI, and CERV-CII. These studies thus identified the first virus from another species that is immunologically related to the MuMTV of M. musculus. Particles similar to MC-MTV were also observed in a spontaneous M. cervicolor popaeus mammary tumor.


Asunto(s)
Virus del Tumor Mamario del Ratón/inmunología , Retroviridae/aislamiento & purificación , Roedores/microbiología , Animales , Antígenos Virales , Epítopos , Femenino , Ratones , Microscopía Electrónica , Leche/microbiología , ADN Polimerasa Dirigida por ARN/metabolismo , Retroviridae/enzimología , Retroviridae/inmunología , Proteínas Virales/inmunología
4.
Cancer Res ; 43(2): 728-35, 1983 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-6848188

RESUMEN

Murine monoclonal antibodies, prepared against human metastatic mammary tumor cells, were used to demonstrate differential expression of several tumor-associated antigens (TAAs) among various mammary carcinomas and within a given tumor mass. Using the immunoperoxidase technique on serial sections of 39 human primary mammary carcinomas, a spectrum of antigenic phenotypes of TAAs was observed: 13% of the tumors reacted with all of a panel of four monoclonal antibodies; while 10% of the mammary tumors scored negative with all four antibodies. The remaining 30 tumors could be divided into several additional groups based on their differential reactivity with some, but not all, of the monoclonal antibodies. Furthermore, variation among mammary carcinomas was also observed in the cellular localization of antigens. Antigenic phenotypic diversity of mammary tumor cell populations within a given tumor mass was also observed; this was noted with respect to (a) antigenic expression in one area of a tumor mass and not another and (b) a "patchwork" effect in which antigens were expressed on cells immediately adjacent to cells which scored negative. Antigenic phenotypic diversity was also observed in established mammary tumor cell lines grown in vitro. A differential loss of some cell surface TAAs was observed as a function of continued cell passage; consistent with this finding, MCF-7 mammary tumor cell lines obtained from four sources could be differentiated from each other by their pattern of cell surface TAA expression. Single-cell clones derived from the MCF-7 mammary tumor cell line exhibited at least four distinct antigenic phenotypes; a change in cell surface phenotype of some of the clones was seen during subsequent passage. This definition of phenotypic variation and modulation of TAA expression among, and within, human mammary carcinomas has implications towards both the design and the outcome of studies involving the in situ immunodetection and therapy of breast cancer.


Asunto(s)
Adenocarcinoma/inmunología , Anticuerpos Monoclonales , Antígenos de Neoplasias/análisis , Neoplasias de la Mama/inmunología , Animales , Línea Celular , Femenino , Humanos , Hibridomas/inmunología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/secundario , Linfocitos/inmunología , Ratones , Plasmacitoma
5.
Cancer Res ; 45(11 Pt 2): 5648-55, 1985 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-2413994

RESUMEN

A new method for the analysis of the binding of monoclonal antibodies to cell surface tumor-associated antigens utilizes 1- to 2-day primary cultures of human colonic carcinomas, adenomas, and normal epithelial tissue. The antibodies are added to the live cells which form monolayer epithelial patches of several hundred cells on the surface of the Petri dish by migration in a continuous sheet from a small explant. These epithelial patches are then fixed with methanol and processed in situ using the indirect immunoperoxidase assay. Three monoclonal antibodies (MAbs) prepared against membrane-enriched fractions of human metastatic breast cancer were assayed. MAb B1.1 bound to each of 11 benign and each of 18 malignant colonic tumors tested. MAb B6.2 displayed similar reactivity, binding to each of 7 adenomas and each of 15 carcinomas assayed. Both MAbs also bound to normal colonic epithelial cells in both the live cell studies presented here and in earlier studies (D. Stramignoni et al., Int. J. Cancer, 31: 543-552, 1983). MAb B72.3 bound only to tumor cells and not to normal epithelial cells in the live cell assay. This epitope was rapidly lost in culture. B72.3 reactivity on each of two carcinomas was decreased 9- to 36-fold when primary culture continued for 5-6 days. B72.3 bound to each of 20 tumors (15 carcinomas, 5 adenomas) when the cells were cultured for 1 or 2 days but on only 2 of 8 tumors when the cells were cultured for 3 to 8 days. The B72.3 epitope was more strongly expressed on the live cells in the explant and on those monolayer cells directly adjacent to the explant than on the cells more towards the edges of the patch colony. This implied that the cell flattening which occurred when cells migrated from the explant may have played some role in antigen loss. A very similar fraction of primary cultured carcinoma and adenoma cells bound each MAb, indicating that these MAbs in live cell assay do not distinguish between benign, noninvasive colonic tumors and invasive carcinomas. The live cell assay was compared to the standard assay utilizing sectioned, fixed tumors. In parallel assays of eight tumors the fraction of cells reactive in the indirect immunoperoxidase assay was consistently higher on live cells for each of these MAbs than on fixed tissue. Due to this greater sensitivity the live cell assay was able to detect reactive cells in two cases which were scored as negative (less than 1% positive cells) in the fixed tissue assay.


Asunto(s)
Antígenos de Neoplasias/análisis , Antígenos de Superficie/análisis , Carcinoma/inmunología , Colon/inmunología , Neoplasias del Colon/inmunología , Adenoma/inmunología , Anticuerpos Monoclonales/inmunología , Células Cultivadas , Epitelio/inmunología , Epítopos/análisis , Humanos
6.
Cancer Res ; 45(6): 2713-9, 1985 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-3157447

RESUMEN

It has been hypothesized that epithelial and endothelial cells interact with the laminin component of basement membranes via a cell surface laminin receptor molecule. It has also been proposed that the expression of this molecule may be involved in the invasion of carcinoma cells from their tissue of origin and their subsequent penetration through blood vessel basement membranes. We report here the use of a monoclonal antibody, LR-3, to define the expression of laminin receptor in normal, dysplastic, and carcinomatous human tissues. Monoclonal antibody LR-3 is shown by immunoblotting to recognize the Mr 67,000 laminin receptor protein, to bind to the carcinoma cells, and to constitute approximately 0.1% of total cellular protein. Numerous normal human epithelial and endothelial cell types, as well as pulmonary macrophages, are shown to express laminin receptor to varying degrees. Selected human mammary carcinomas and colon carcinomas are shown to bind more monoclonal antibody LR-3 than normal or dysplastic counterparts. A monoclonal antibody to laminin receptor now makes possible the study of the role of laminin receptor in tumor cell metastases and in the differentiation and function of various normal human epithelial and endothelial cell types.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Neoplasias/análisis , Receptores Inmunológicos/análisis , Animales , Mama/análisis , Neoplasias de la Mama/análisis , Colon/análisis , Neoplasias del Colon/análisis , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Receptores Inmunológicos/inmunología , Receptores de Laminina , Piel/análisis
7.
Cancer Res ; 55(11): 2455-62, 1995 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-7538903

RESUMEN

Human prostate-specific antigen (PSA) has been shown as an aid in the early detection of prostate cancer (W. J. Catalona et al., J. Am. Med. Assoc., 270: 948-954, 1993) and was approved in 1994 by the Food and Drug Administration for early detection of prostate cancer. Immunotherapies directed against PSA have been suggested in patients with metastatic prostate cancer. One of the essential questions is to define which nonhuman species express PSA for experimental studies. Using Southern blot analyses, genes related to human PSA have been detected in several nonhuman primate species, including chimpanzee, orangutan, gorilla, macaque, and rhesus monkey, but not in other mammalian species, including rabbit, cow, pig, dog, rat, or mouse. Immunohistochemical staining with anti-human PSA antisera detected strong staining in both human and monkey prostatic epithelial cells with no reactivity to rat prostate cells. Because the PSA gene is not present in the murine genome, a matched set of murine cell lines has been developed that may be useful to study the biochemical functions of PSA and as an experimental target for PSA-directed immunotherapy. To establish such cell lines, a C57BL/6 murine colon adenocarcinoma cell line, MC-38, was transfected with a retroviral vector containing cDNA encoding the human PSA gene. Genetic analysis of a PSA-secreting clone, PSA/MC-38, demonstrated that the PSA gene had been stably integrated into the MC-38 genome. The PSA/MC-38 cell line was found to secrete PSA into tissue culture medium, producing a protein of approximately M(r) 30,000. In vivo, PSA/MC-38 grew as a s.c. tumor in male and female mice. PSA/MC-38 tumors grew more rapidly in athymic mice than in syngeneic C57BL/6 mice, and in both mouse strains, the PSA/MC-38 tumors grew more slowly than control vector-transduced tumors. PSA was detected in the serum and tumors of PSA/MC-38 tumor-bearing mice. It is proposed that PSA/MC-38 cells may be used as a murine tumor model to test potential therapeutic vaccines and other experimental therapies directed against PSA.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Antígeno Prostático Específico/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/química , Animales , Secuencia de Bases , División Celular/fisiología , Neoplasias del Colon/química , ADN de Neoplasias/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Genoma , Humanos , Inmunohistoquímica , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Datos de Secuencia Molecular , Próstata/química , Próstata/metabolismo , Próstata/fisiología , Antígeno Prostático Específico/análisis , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/química , Ratas , Ratas Wistar , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección , Células Tumorales Cultivadas
8.
Cancer Res ; 51(14): 3657-62, 1991 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-1712245

RESUMEN

A cell line derived from the mouse colon adenocarcinoma, MC-38, has been transduced with a retroviral construct containing complementary DNA encoding the human carcinoembryonic antigen (CEA) gene. MC-38, which forms tumors in syngeneic C57BL/6 mice, has been extensively studied as a target for active immunotherapy. Individual transduced clones that express high levels of cell surface CEA were isolated, and two clones, termed MC-38-ceal and MC-38-cea2, were extensively characterized. The levels of CEA found on the surface of these clones were considerably higher than that found in a moderately differentiated human colon carcinoma cell line (WiDr) and were comparable to those found on the human colon carcinoma cell lines GEO and CBS (among the highest CEA-expressing cells reported). Further analysis demonstrated that the CEA expressed in the MC-38-cea1 clone had a similar molecular weight to native CEA (Mr 180,000), but the MC-38-cea2 cell line expressed a single Mr 70,000 glycosylated immunoreactive product. Seven anti-CEA monoclonal antibodies were found to react with both clones. The CEA gene present in the MC-38-cea2 clone was partially sequenced and was found to contain a deletion of two of the three repeated domains present in CEA. These results provide a basis for future studies to map immunodominant epitopes of CEA and to develop a syngeneic model system that may aid in the design of reagents and protocols to study active and passive immunotherapy directed against a carcinoma expressing human CEA.


Asunto(s)
Adenocarcinoma/inmunología , Antígenos de Neoplasias , Antígeno Carcinoembrionario/genética , Moléculas de Adhesión Celular , Neoplasias del Colon/inmunología , Expresión Génica , Transducción Genética , Adenocarcinoma/genética , Animales , Secuencia de Bases , Antígeno Carcinoembrionario/análisis , Antígeno Carcinoembrionario/inmunología , Neoplasias del Colon/genética , Epítopos/análisis , Glicoproteínas de Membrana/inmunología , Ratones , Datos de Secuencia Molecular , Peso Molecular , Células Tumorales Cultivadas
9.
Cancer Res ; 44(8): 3208-14, 1984 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6744259

RESUMEN

Treatment of human breast or colon carcinoma cells with recombinantly derived human leukocyte (clone A) interferon (IFN-alpha A) increases the surface expression of specific tumor-associated antigens (TAAs) recognized by monoclonal antibodies (MAbs). The MAbs used, B1.1, B6.2, and B72.3, recognize three distinct TAAs, i.e., the Mr 180,000 carcinoembryonic antigen, a Mr 90,000, and a Mr 220,000 to 400,000 glycoprotein, respectively. The binding of the MAbs to the surface of tumor cells increased in a dose-dependent manner, with optimal levels of TAA enhancement at 100 to 1,000 units IFN-alpha A/ml. Higher concentrations of IFN-alpha A that were cytostatic or cytotoxic were also less effective in enhancing TAA expression. Human melanoma (A375) cells and normal fibroblasts (WI-38 and Flow 4000) do not express any of the three TAAs, either before or after interferon treatment. The ability of IFN-alpha A to increase the expression of TAAs on human carcinoma cells was also temporally dependent, with optimal enhancement occurring after 16 to 24 hr. The enhancement of specific TAAs at the surface of the carcinoma cells by IFN-alpha A was confirmed, using fluorescence-activated cell sorter analysis. These data demonstrate that the IFN-alpha A-mediated increase of surface antigen is a result of both an accumulation of more antigen per cell, and an increase in the percentage of cells expressing the antigen. The ability of recombinant interferon to enhance specific TAAs on human carcinoma cells may be exploited in designing protocols for the in situ detection and therapy of human carcinoma lesions by MAbs, as well as in further defining the role of specific TAAs in the expression of the transformed phenotype.


Asunto(s)
Antígenos de Neoplasias/aislamiento & purificación , Antígenos de Superficie/aislamiento & purificación , Neoplasias de la Mama/inmunología , Neoplasias del Colon/inmunología , Interferón Tipo I/inmunología , Anticuerpos Monoclonales , Antígenos de Neoplasias/genética , Antígenos de Superficie/genética , Antígeno Carcinoembrionario/aislamiento & purificación , Femenino , Citometría de Flujo , Humanos , Interferón Tipo I/genética , Peso Molecular , Radioinmunoensayo
10.
Cancer Res ; 46(5): 2511-9, 1986 May.
Artículo en Inglés | MEDLINE | ID: mdl-3084069

RESUMEN

Monoclonal antibodies RAP-5 and Y13-259, directed against the ras gene product [a protein with a molecular weight of 21,000 (p21)] have been used to evaluate ras p21 expression in malignant and benign mammary tissues as well as in the lesions of intermediate stature such as atypical hyperplasia using immunohistochemical assays. Invasive carcinoma demonstrated enhanced expression of ras p21, with generally decreasing expression in carcinoma in situ, atypical hyperplasia, and nonatypical hyperplasia, respectively. Heterogeneous expression of ras p21 was observed among primary as well as metastatic mammary carcinomas. Carcinomas from postmenopausal patients generally demonstrated higher levels of ras p21 than those from premenopausal patients, but no significant difference in ras p21 expression in carcinomas between estrogen-receptor rich and estrogen-receptor poor patients was found. Normal mammary epithelium in terminal duct lobular units from patients with hyperplasia generally demonstrated higher levels of ras p21 expression than did epithelium in large ducts. This demonstration of enhanced ras p21 expression by the epithelium of peripheral lobular portion of the breast is consistent with the previous hypothesis that these areas preferentially undergo malignant transformation. Analyses of the limited number of specimens available from patients with 15-yr follow-up revealed a generally higher level of ras p21 in hyperplasia from patients who subsequently developed carcinoma, as compared to those from patients without carcinoma development. However, no conclusions regarding the potential for malignant transformation could be drawn for any individual patient on the basis of ras p21 expression. Concomitant analyses of ras p21 expression in mammary carcinomas and benign lesions using liquid competition radioimmunoassay and immunohistochemical assay demonstrated the complementary nature of these alternative approaches. These results suggest that enhanced ras p21 expression may be involved in the early stages of mammary carcinogenesis but is probably not involved in the maintenance of the transformed phenotype.


Asunto(s)
Enfermedades de la Mama/metabolismo , Neoplasias de la Mama/metabolismo , Mama/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Anticuerpos Monoclonales , Carcinoma/metabolismo , Carcinoma in Situ/metabolismo , Epitelio/metabolismo , Epitelio/patología , Femenino , Enfermedad Fibroquística de la Mama/metabolismo , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/inmunología , Humanos , Hiperplasia , Técnicas para Inmunoenzimas , Menopausia , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Receptores de Estrógenos/metabolismo
11.
Cancer Res ; 51(1): 181-9, 1991 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-1988082

RESUMEN

We report here the generation and characterization of a recombinant/chimeric construct of murine gamma 1 monoclonal antibody (MAb) B72.3, containing the murine variable region and a human gamma 1 constant region [designated cB72.3(gamma i)]. cB72.3(gamma 1) was generated by first isolating functionally rearranged VH and VL genes of B72.3 from partial genomic libraries in phage vectors. Construction of mouse-human chimeric heavy and light chain genes was performed by inserting restriction fragments carrying VL and VH regions of B72.3 into unique sites of expression vectors which contains sequences encoding constant regions of human kappa and gamma 1, respectively. The expression constructs were subsequently electroporated into SP2/0 cells. The transfected SP2/0 murine cell line has been shown to synthesize cB72.3(gamma 1) at a level of 10-20 micrograms/ml. Reciprocal competition radioimmunoassays demonstrated that cB72.3(gamma 1), a previously described cB72.3(gamma 4), and native B72.3 (designated nB72.3) competed similarly. A rat anti-idiotype MAb made against nB72.3 was shown to bind equally well to cB72.3(gamma 1) and to the nB72.3. Immunochemical studies of the nB72.3, cB72.3(gamma 4), and cB72.3(gamma 1) revealed slight differences in size among the three MAb forms on sodium dodecyl sulfate gels and revealed a higher isoelectric point for the cB72.3(gamma 1). Antibody-dependent cell-mediated cytotoxicity experiments using human lymphokine-activated killer effector cells indicated better tumor cell killing by the cB72.3(gamma 1) than the nB72.3 or cB72.3(gamma 4). Dual label studies of coinjected cB72.3(gamma 1) and nB72.3 revealed that both MAbs could efficiently localize human tumor xenografts in athymic mice. Pharmacokinetic studies, analyzing the blood clearance of cB72.3(gamma 1), cB72.3(gamma 4), and nB72.3 in mice, showed that the nB72.3 beta phase of clearance was slower than that of other MAb forms. However, when the pharmacokinetic patterns of these three MAbs forms were analyzed in monkeys, the cB72.3(gamma 1) and the nB72.3 showed similar clearance curves, while the cB72.3(gamma 4) showed a much slower plasma clearance. In view of the binding properties of nB72.3 and its ability to localize a range of carcinomas in clinical trials, the studies reported here demonstrate that the cB72.3(gamma 1) may serve as a potentially useful diagnostic and/or therapeutic reagent.


Asunto(s)
Anticuerpos Monoclonales/genética , Anticuerpos Antineoplásicos/genética , Carcinoma/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/farmacocinética , Especificidad de Anticuerpos , Citotoxicidad Celular Dependiente de Anticuerpos , Secuencia de Bases , Unión Competitiva , Quimera , Clonación Molecular , Reordenamiento Génico de Linfocito B , Genes de Inmunoglobulinas , Vectores Genéticos , Punto Isoeléctrico , Macaca fascicularis , Ratones , Ratones Desnudos , Proteínas Recombinantes de Fusión , Transfección
12.
Cancer Res ; 47(5): 1413-20, 1987 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-2434216

RESUMEN

Using c-Ha-, c-Ki-, and c-N-ras-specific probes in a RNA-RNA hybridization assay we found enhanced expression of c-Ha-ras protooncogene in stomach adenocarcinomas relative to nonneoplastic epithelium, whereas little or no transcription of either c-Ki- or c-N-ras was detected. Enhanced levels of c-Ha-ras RNA expression were detected in all of the adenocarcinomas examined. Hybridization with c-Ha-ras was also detected in nonneoplastic gastric epithelium adjacent to carcinoma, although the labeling was less intense than that of carcinoma cells. More extensive analysis of the c-Ha-ras p21 expression was then carried out in formalin-fixed, paraffin-embedded tissue sections and extracts from surgically resected stomach tissues using monoclonal antibodies (MAbs) RAP-5 and Y13-259. The data obtained from the immunohistochemical studies were consistent with the results of in situ hybridization assay. Adenocarcinomas were much more reactive with MAb RAP-5 than benign and normal tissues, and the majority of carcinomas demonstrated increased expression of c-Ha-ras p21. Quantitative liquid competition radioimmunoassays using MAb Y13-259 also demonstrated significantly higher levels of c-Ha-ras p21 in extracts from stomach adenocarcinomas than those from normal mucosae. No strict correlation was found between ras p21 expression and the degree of tumor differentiation or histological type. Although advanced carcinomas generally demonstrated higher levels of ras p21 than early carcinomas, no correlation among advanced carcinomas and ras p21 levels was observed in relation to depth of tumor invasion to the muscularis propria, subserosa, or serosa. Benign lesions, in comparison, were much less reactive with MAb RAP-5 than carcinomas. Among the benign lesions tested, dysplastic lesions were more reactive than nondysplastic lesions. Normal stomach mucosa was generally nonreactive with the exception of parietal cells. Our results indicate that transformation of the stomach mucosa from benign to malignant phenotype is associated with an increase in c-Ha-ras p21 expression.


Asunto(s)
Adenocarcinoma/análisis , Anticuerpos Monoclonales/inmunología , Proteínas Proto-Oncogénicas/análisis , Neoplasias Gástricas/análisis , Mucosa Gástrica/análisis , Histocitoquímica , Humanos , Hibridación de Ácido Nucleico , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas p21(ras) , Proto-Oncogenes , ARN/análisis , Radioinmunoensayo
13.
Cancer Res ; 50(3 Suppl): 820s-827s, 1990 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-1688734

RESUMEN

Tumor targeting by monoclonal antibodies (MAbs) can be enhanced by (a) increasing the percentage of injected dose taken up by the tumor and/or (b) increasing the tumor:nontumor ratios. Several groups have demonstrated that one can increase tumor to nontumor ratios by the use of antibody fragments or the administration of second antibodies. Several other modalities are also possible: (a) the use of recombinant interferons to up-regulate the expression of specific tumor associated antigens such as carcinoembryonic antigen or TAG-72 on the surface of carcinoma cells and thus increase MAb tumor binding has proved successful in both in vitro and in vivo studies; (b) the intracavitary administration of MAbs. Recent studies have demonstrated that when radiolabeled B72.3 is administered i.p. to patients with carcinoma of the peritoneal cavity, it localizes tumor masses with greater efficiency than does concurrent i.v. administered antibody. Studies involving the comparative pharmacology of intracavitary administration of radiolabeled MAb in patients and several animal models will be discussed; (c) it has been reported that prior exposure of hepatoma to external beam radiation will increase radiolabeled MAb tumor targeting. We and others have not been able to duplicate this phenomenon with a human colon cancer xenograft model and radiolabeled MAbs to two different colon carcinoma associated antigens. The possible reasons for these differences will be discussed; (d) the cloning and expression of recombinant MAbs with human constant regions and subsequent size modification constructs will also undoubtedly alter the pharmacology of MAb tumor binding in both diagnostic and therapeutic applications.


Asunto(s)
Anticuerpos Monoclonales , Neoplasias/diagnóstico , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos de Neoplasias/biosíntesis , Humanos , Interferones/farmacología , Radioisótopos de Yodo/administración & dosificación , Proteínas Recombinantes
14.
Clin Cancer Res ; 5(10 Suppl): 3118s-3123s, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10541352

RESUMEN

Monoclonal antibody (mAb) CC49, a murine IgG1, reacts with the tumor-associated glycoprotein-72 expressed on a variety of carcinomas. In clinical trials, radiolabeled CC49 has shown excellent tumor localization to a variety of carcinomas. To minimize the immunogenicity of CC49 mAb in patients, a humanized CC49 (HuCC49) was generated by complementarity-determining region (CDR) grafting. The relative affinity of HuCC49 was 2-3-fold less than that of the murine mAb. With the aim of improving tumor targeting, attempts have been made to enhance the avidity of the HuCC49 mAb. Previous research has yielded a single gene-encoded immunoglobulin, SCIgcCC49deltaCH1, which is a dimer of a single chain consisting of CC49 single-chain Fv linked to the NH2 terminus of the human gamma1 Fc through the hinge region. This molecule is comparable to the mouse-human chimeric CC49 in terms of in vitro antigen binding properties, cytolytic activity, and rate of plasma clearance in athymic mice bearing human tumor xenografts. Recently, a single gene encoding a single-chain immunoglobulin consisting of a HuCC49 diabody attached to human gamma1 Fc via the hinge region was constructed. The diabody, a bivalent antigen-binding structure, is made up of variable heavy (V(H))/variable light (V(L)) domains and V(L)/V(H) domains. In each of the variable domain pairs, the V(H) and V(L) domains are linked through a short linker peptide. Meanwhile, the two pairs are linked via a 30-residue Gly-Ser linker peptide to yield two antigen-binding sites by lateral and noncovalent association of the V(L) of one pair with the V(H) of the other. Transfectomas expressing the single-gene immunoglobulin secrete a homodimer of about Mr 160,000 that reacts to tumor-associated glycoprotein-72. This tetravalent humanized antitumor immunoglobulin molecule may potentially be an efficacious therapeutic and diagnostic reagent against a wide range of human carcinomas.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Antígenos de Neoplasias/inmunología , Glicoproteínas/inmunología , Inmunoglobulina G/biosíntesis , Neoplasias/terapia , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Unión Competitiva , Humanos , Ratones
15.
Mol Immunol ; 36(1): 61-71, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10369421

RESUMEN

Monoclonal antibody (MAb) CC49 is a murine IgG1 that reacts with tumor-associated glycoprotein (TAG)-72, a pancarcinoma antigen. Clinical trials using radiolabeled CC49 for diagnostic imaging have demonstrated specific localization of more than 90% of carcinomas. The feasibility of adopting in vivo gene inoculation methods for antibody-based immunotherapy requires introduction and expression of two genes, encoding immunoglobulin (Ig) heavy and light chains, in a single cell to generate a functional antibody. To circumvent the problems inherent in this approach, we have constructed a single-gene encoding a single-chain immunoglobulin (SCIg) that, unlike previously developed SCIgs, contains all IgG domains. To construct the novel SCIg, the carboxyl end of the constant region of the chimeric (c) CC49 kappa chain is joined, via a 30 residue Gly-Ser linker peptide, to the amino terminus of the CC49 heavy chain. To our knowledge, neither a linker peptide this long nor a linkage between the constant light (C(L)) and variable heavy domains has been reported previously. Transfectomas developed by introducing the expression construct of the amplifiable gene in dihydrofolate reductase-deficient Chinese hamster ovary (CHO dhfr-) cells secrete a 160 kDa homodimeric molecule, SCIgcCC49. The in vitro antigen binding properties of SCIgcCC49 are comparable to those of cCC49 and SCIgcCC49deltaC(H)1, a single-chain Ig deficient in constant heavy chain-1 (C(H)1) and C(L) domains. The antibody-dependent cellular cytotoxicity (ADCC) of SCIgcCC49 and cCC49 were also comparable. This single-gene approach for generating an immunoglobulin molecule may facilitate in vivo gene inoculation as well as ex vivo transfection of patients' cultured tumor-infiltrating lymphocytes for immunotherapy protocols for a variety of diseases, including cancer.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Reacciones Antígeno-Anticuerpo , Genes de Inmunoglobulinas , Cadenas Pesadas de Inmunoglobulina/inmunología , Cadenas Ligeras de Inmunoglobulina/inmunología , Cadenas kappa de Inmunoglobulina/inmunología , Animales , Anticuerpos Monoclonales/genética , Especificidad de Anticuerpos , Sitios de Unión/inmunología , Cricetinae , Ensayo de Inmunoadsorción Enzimática , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/genética , Ratones
16.
Hum Gene Ther ; 10(2): 165-73, 1999 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-10022542

RESUMEN

A chimeric immune receptor consisting of an extracellular antigen-binding domain derived from the CC49 humanized single-chain antibody, linked to the CD3zeta signaling domain of the T cell receptor, was generated (CC49-zeta). This receptor binds to TAG-72, a mucin antigen expressed by most human adenocarcinomas. CC49-zeta was expressed in CD4+ and CD8+ T cells and induced cytokine production on stimulation. Human T cells expressing CC49-zeta recognized and killed tumor cell lines and primary tumor cells expressing TAG-72. CC49-zeta T cells did not mediate bystander killing of TAG-72-negative cells. In addition, CC49-zeta T cells not only killed FasL-positive tumor cells in vitro and in vivo, but also survived in their presence, and were immunoprotective in intraperitoneal and subcutaneous murine tumor xenograft models with TAG-72-positive human tumor cells. Finally, receptor-positive T cells were still effective in killing TAG-72-positive targets in the presence of physiological levels of soluble TAG-72, and did not induce killing of TAG-72-negative cells under the same conditions. This approach is being currently being utilized in a phase I clinical trial for the treatment of colon cancer.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptores Inmunológicos/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Antineoplásicos/genética , Antígenos de Neoplasias/inmunología , División Celular/inmunología , Proteína Ligando Fas , Glicoproteínas/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/citología , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/citología
17.
Semin Oncol ; 23(1): 118-34, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8607022

RESUMEN

The ras p21 proto-oncogenes (ie, K-ras, H-ras, N-ras) encode a family of proteins vital to cellular signaling and function. Point mutations in these genes have been found in a wide diversity of human cancers, suggesting a strong association in the development of the malignant phenotype. Although the precise mechanisms leading to tumorigenesis are not fully understood, it has been proposed that point mutations in the ras p21 proto-oncogenes contribute to the transformation process through constitutive transduction of growth-promoting signals. These oncoproteins are distinct from normal ras p21 in both DNA and protein sequences at specific sites, typically positions 12, 13, 59, or 61. A large frequency of human cancers harbor point mutations in the ras gene at codon 12, where the normal Gly residue is substituted with either a Val, Asp or Cys residue. From an immunologic perspective, these "neo-determinants" may now represent unique and highly specific epitopes for T cell (CD4+ and/or CD8+) recognition in cancer immunotherapy. Evaluation of point-mutated ras as a T-cell epitope could be determined biologically with short synthetic peptides that precisely mimic those altered sites. Several laboratories have established approaches in both murine and human systems to evaluate the point-mutated ras p21 oncogene product as a potential tumor-specific target and characterization of the resulting cellular immune responses. It has been demonstrated that (1) active immunization of mice with the appropriate mutant protein or peptides leads to the production of cytotoxic CD4+ (Th1 subtype) or CD8+ T lymphocytes, which mediate MHC-restricted, antigen-specific lysis of tumor cells in vitro bearing endogenous mutant ras epitopes; and (2) in vitro stimulation of human lymphocytes from some normal individuals or carcinoma patients with mutant ras peptides results in the expansion of CD4+ and CD8+ precursors, which may exhibit cytotoxicity against autologous or MHC-matched, antigen-bearing target cells. Taken collectively, these preclinical findings provide the rationale for the development of potential immunotherapies directed against point-mutated ras oncogene products.


Asunto(s)
Epítopos/inmunología , Neoplasias/inmunología , Mutación Puntual , Proteínas Proto-Oncogénicas p21(ras)/inmunología , Vacunas/inmunología , Animales , Humanos , Inmunoterapia Activa/métodos , Ratones , Linfocitos T Citotóxicos/inmunología
18.
Int J Radiat Oncol Biol Phys ; 16(3): 721-9, 1989 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2921170

RESUMEN

Recent studies in animal models involving antibody tumor targeting of hepatoma and melanoma and clinical trials involving hepatoma patients have suggested that preirradiation of tumors may enhance antibody tumor targeting. These reports led us to study the effect of external irradiation on monoclonal antibody (MAb) targeting of human carcinomas; as a model system, we used MAb B72.3 and the LS-174T human colon carcinoma xenograft in athymic mice. LS-174T tumors exposed to 300 cGy grew to approximately 93% the size of non-irradiated tumors, while those exposed to 600, 900, or 2,000 cGy were approximately 41% the size of control tumors. Splitting the 900 cGy into three 300-cGy fractions yielded a two-fold lower tumor volume compared with a single 900-cGy fraction. Histochemical evaluation of the carcinomas revealed a decrease in the number of mitoses per high power field consistent with early effects of radiation exposure. Using the avidin-biotin complex immunoperoxidase technique, carcinomas were assayed for expression of the tumor associated glycoprotein (TAG)-72, the high-molecular-weight mucin detected by MAb B72.3. No discernable variation was observed in the staining intensity among tumors in both the control and radiation treated group; that is, differences among tumors within each group were compatible with the known heterogeneous expression of TAG-72. Exposure of carcinomas to 300 or 900 cGy in a single fraction or 900 cGy split in three 300-cGy fractions did not yield a consistent or substantial enhanced localization of radiolabeled MAb B72.3 IgG or F(ab')2 to tumors. A 1.5-fold augmentation of MAb binding to tumors was observed in preirradiated mice; however, these results were not statistically significant. Inherent differences in tumors such as cell type of origin, size, spatial configuration, extent of vascularization and volume of interstitial space may contribute to variability of the effect of preirradiation of tumors on antibody binding. Our results suggest that consistent augmentation of radiolabeled antibody localization to tumors is not a universal phenomenon.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/inmunología , Neoplasias del Colon/radioterapia , Glicoproteínas/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Neoplasias del Colon/diagnóstico por imagen , Neoplasias del Colon/inmunología , Femenino , Humanos , Radioisótopos de Yodo/uso terapéutico , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Cintigrafía , Trasplante Heterólogo
19.
Cancer Treat Res ; 51: 413-32, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-1977458

RESUMEN

The use of Mabs for the detection and treatment of human carcinoma lesions can still be regarded in its infancy. As with other new approaches to cancer therapy, several conceptual as well as real problems exist when designing clinical protocols for Mab-directed immunotherapy. From the Mab standpoint, studies using the intact IgG have shown that, in a majority of patients injected with IgG, human anti-mouse IgG antibodies develop that hamper the effectiveness of subsequent antibody administration. It is believed that the human anti-mouse antibody response is directed against the Fc region of the IgG molecule. The elimination of this region through fractionation of the Mab to obtain the minimum binding site could result in a less immunogenic molecule. Another approach aimed at reducing the immunogenicity of the Mab would be to clone the genes encoding for individual Mabs, reduce them via restriction endonuclease techniques, and insert human immunoglobulin constant regions. The resulting chimeric antibodies are believed to reduce the development of human anti-mouse antibodies. Effective Mab therapy of human tumor lesions may also be achieved through the recruitment of a portion of the host's immunologic defense system. An example is the use of anti-idiotype Mabs that use as immunogen a Mab to a tumor antigen. The anti-idiotype antibodies are selected for binding to the antigen binding, or idiotype, region of the first Mab. The binding sites of the new anti-idiotype Mabs should reflect the 'internal image' of the original antigen. The anti-idiotype antibodies may be used to immunize patients (i.e., vaccines) in an attempt to mount an active immune response against the antigen-positive tumor cells. Recent studies have shown a synergism between interferon-alpha and an anti-idiotype Mab for the in-vivo antitumor activity in a murine B-cell lymphoma experimental model. Whether an interferon-mediated increase in the tumor antigen or the Fc receptor was part of the synergism was not investigated. Mabs alone have also been shown to elicit cytotoxic activity in vitro and tumoricidal activity in vivo. Antibodies of the IgG2a isotype can direct macrophage-mediated cytotoxicity. These studies revealed the importance of the number of antibody sites per cell as well as the number of cells that bind the IgG2a Mab, thus suggesting a 'threshold' requirement for the demonstration of effective tumor cell lysis in vitro and in vivo.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Anticuerpos Monoclonales , Antígenos de Neoplasias/análisis , Interferón Tipo I/farmacología , Interferón gamma/farmacología , Neoplasias/diagnóstico por imagen , Antígeno Carcinoembrionario/análisis , Glicoproteínas/análisis , Humanos , Cintigrafía , Proteínas Recombinantes
20.
Anticancer Res ; 11(4): 1657-62, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1660693

RESUMEN

A monoclonal antibody, RAP-5, raised against the 21,000d ras-oncogene product, p21, was used in an immunoperoxidase staining procedure to study the expression of p21 in normal children's and foetal kidneys, pre-malignant lesions and benign and malignant childhood renal tumours with good, moderate or poor prognosis. ras-p21 was expressed in both normal and foetal kidneys but its distribution in renal tumours differed markedly (p less than 0.01). A quantitative liquid competition radioimmunoassay (RIA) was used to determine ras-p21 level in tissue homogenates. The results were expressed as pg of ras-p21 per microgram protein of tissue extract. There were significant differences in the levels of ras-p21 among various renal tissue extracts (p less than 0.05). Generally it emerged that the amount of ras-p21 was greater in both malignant renal tumours and foetal kidneys compared to normal kidneys, pre-malignant lesions and benign renal tumours.


Asunto(s)
Biomarcadores de Tumor/análisis , Genes ras , Neoplasias Renales/patología , Riñón/fisiología , Proteínas Proto-Oncogénicas p21(ras)/análisis , Anticuerpos Monoclonales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Niño , Feto , Humanos , Técnicas para Inmunoenzimas , Riñón/citología , Riñón/embriología , Neoplasias Renales/genética , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Radioinmunoensayo , Valores de Referencia , Tumor de Wilms/genética , Tumor de Wilms/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA