Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Skin Health Dis ; 1(1)2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34541577

RESUMEN

BACKGROUNDS: Folate Hydrolase-1 (FOLH1; PSMA) is a type II transmembrane protein, luminally expressed by solid tumour neo-vasculature. Monoclonal antibody (mAb), J591, is a vehicle for mAb-based brachytherapy in FOLH1+ cancers. Brachytherapy is a form of radiotherapy that involves placing a radioactive material a short distance from the target tissue (e.g., on the skin or internally); brachytherapy is commonly accomplished with the use of catheters, needles, metal seeds and antibody or small peptide conjugates. Herein, FOLH1 expression in primary (p) and metastatic (m) Merkel cell carcinoma (MCC) is characterized to determine its targeting potential for J591-brachytherapy. MATERIALS & METHODS: Paraffin sections from pMCC and mMCC were evaluated by immunohistochemistry for FOLH1. Monte Carlo simulation was performed using the physical properties of conjugated radioisotope lutetium-177. Kaplan-Meier survival curves were calculated based on patient outcome data and FOLH1 expression. RESULTS: Eighty-one MCC tumours were evaluated. 67% (54/81) of all cases, 77% (24/31) pMCC and 60% (30/50) mMCC tumours were FOLH1+. Monte Carlo simulation showed highly localized ionizing tracks of electrons emitted from the targeted neo-vessel. 42% (34/81) of patients with FOLH1+/- MCC had available survival data f or analysis. No significant differences in our limited data set were detected based on FOLH1 status (p = 0.4718; p = 0.6470), staining intensity score (p = 0.6966; p = 0.9841) or by grouping staining intensity scores (- and + vs. ++, +++, +++) (p = 0.8022; p = 0.8496) for MCC-specific survival or recurrence free survival, respectively. CONCLUSIONS: We report the first evidence of prevalent FOLH1 expression within MCC-associated neo-vessels, in 60-77% of patients in a large MCC cohort. Given this data, and the need for alternatives to immune therapies it is appropriate to explore the safety and efficacy o f FOLH1-targeted brachytherapy for MCC.

2.
J Exp Med ; 169(3): 953-72, 1989 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-2494294

RESUMEN

Normal human kidney proximal tubule cells into which a ras oncogene was inserted undergo a series of transformation-related alterations that are characteristic of renal carcinomas. These include changes in morphology, growth potential, anchorage dependence, antigen expression, growth factor production, and chromosomal stability. Further, there are spontaneous progressive alterations in vitro in the karyotype and antigenic profile of the transformed cells. Cytogenetic analyses suggest that alterations of chromosome 21 may play an early and pivotal role in the development of transformed proximal tubule cells.


Asunto(s)
Transformación Celular Neoplásica , Transformación Celular Viral , Genes ras , Neoplasias Renales , Túbulos Renales Proximales , Retroviridae/genética , Antígenos de Neoplasias/análisis , Antígenos de Superficie/análisis , División Celular , Línea Celular Transformada , Cromosomas Humanos Par 21 , Gangliósidos/inmunología , Sustancias de Crecimiento/biosíntesis , Humanos , Cariotipificación , Neoplasias Renales/genética , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Túbulos Renales Proximales/inmunología , Túbulos Renales Proximales/patología , Fenotipo , Células Tumorales Cultivadas
3.
Science ; 294(5546): 1537-40, 2001 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-11711678

RESUMEN

A single, high linear energy transfer alpha particle can kill a target cell. We have developed methods to target molecular-sized generators of alpha-emitting isotope cascades to the inside of cancer cells using actinium-225 coupled to internalizing monoclonal antibodies. In vitro, these constructs specifically killed leukemia, lymphoma, breast, ovarian, neuroblastoma, and prostate cancer cells at becquerel (picocurie) levels. Injection of single doses of the constructs at kilobecquerel (nanocurie) levels into mice bearing solid prostate carcinoma or disseminated human lymphoma induced tumor regression and prolonged survival, without toxicity, in a substantial fraction of animals. Nanogenerators targeting a wide variety of cancers may be possible.


Asunto(s)
Actinio/uso terapéutico , Inmunoconjugados/uso terapéutico , Neoplasias/radioterapia , Radioinmunoterapia/métodos , Actinio/administración & dosificación , Actinio/farmacocinética , Partículas alfa/uso terapéutico , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Femenino , Semivida , Compuestos Heterocíclicos con 1 Anillo , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/farmacocinética , Transferencia Lineal de Energía , Linfoma/radioterapia , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/radioterapia , Tasa de Supervivencia , Células Tumorales Cultivadas
4.
J Clin Invest ; 81(3): 818-21, 1988 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3343341

RESUMEN

A number of factors have been proposed as potential mediators of the syndrome of humoral hypercalcemia of malignancy (HHM), but to date no firm cause-and-effect relationship has been established. We attempted to establish such a relationship by determining whether the presence or absence of adenylate cyclase-stimulating activity (ACSA) in the media of cultured tumor cells predicted the occurrence of the syndrome of HHM when these cell lines were grown in nude mice in vivo. Conditioned media from 35 human renal carcinoma cell lines were surveyed for ACSA in the PTH-sensitive rat osteosarcoma 17/2.8 cell assay. 12 lines were positive (mean, 13.7-fold stimulation, range, 3.0 to 44.0), and 23 lines were negative (mean, 1.2-fold stimulation, range, 0.9 to 1.5). We were successful in establishing five of the positive and six of the negative lines in three to five nude mice per line. Mice implanted with the positive lines uniformly became hypercalcemic (mean serum calcium, 15.8 mg/dl), whereas mice implanted with the negative lines uniformly remained normocalcemic (mean serum calcium, 9.5 mg/dl), in spite of comparable mean tumor size. Acid-urea tumor extracts from each of four hypercalcemic animals contained potent in vitro ACSA (mean, 15.9-fold stimulation), while 5/5 extracts from normocalcemic animals did not (mean, 1.4-fold stimulation). Our study demonstrates that in this model system in vitro ACSA is a reliable predictive marker for HHM in vivo. Whether the protein responsible for this activity is also the mediator of the bone resorption seen in HHM remains to be demonstrated.


Asunto(s)
Adenilil Ciclasas/metabolismo , Hipercalcemia/enzimología , Neoplasias Experimentales/enzimología , Animales , Carcinoma/enzimología , Carcinoma/patología , División Celular , Línea Celular , Medios de Cultivo/análisis , Activación Enzimática , Humanos , Hipercalcemia/patología , Neoplasias Renales/enzimología , Neoplasias Renales/patología , Ratones , Ratones Desnudos , Neoplasias Experimentales/patología , Células Tumorales Cultivadas
5.
J Clin Invest ; 81(6): 2010-4, 1988 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-2454953

RESUMEN

The syndrome of humoral hypercalcemia of malignancy (HHM) appears to be mediated in many instances by a parathyroid hormone-like peptide, which has recently been purified, sequenced, and cloned. Using a probe representing the coding region of the human PTH-like peptide, we examined by Northern analysis poly (A)+ RNA from a variety of human and animal tumors associated with HHM. Hybridizing transcripts were identified in mRNA from each of 12 human and each of four animal HHM-associated tumors, with a complex hybridization pattern observed in the human mRNAs and a relatively simple pattern observed in the animal mRNAs. Poly (A)+ RNA prepared from tumors of similar histological types unassociated with HHM failed to hybridize with the probe. Messenger RNA-dependent biological activity from the animal tumors was entirely eliminated in a hybridization-arrest experiment using a complementary oligonucleotide spanning the region of homology between human PTH and the PTH-like peptide. These findings indicate that the PTH-like peptide is associated with the syndrome of HHM in a wide spectrum of tumor types from a variety of mammalian species and that the PTH-like sequence in the proximal amino terminus of the peptide is highly conserved.


Asunto(s)
Hipercalcemia/genética , Proteínas de Neoplasias/genética , Síndromes Paraneoplásicos/genética , Hormona Paratiroidea/genética , ARN Mensajero/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Humanos , Neoplasias Renales/genética , Datos de Secuencia Molecular , Hibridación de Ácido Nucleico , Proteína Relacionada con la Hormona Paratiroidea , Poli A/genética , ARN/genética , ARN Neoplásico/genética , Homología de Secuencia de Ácido Nucleico , Transcripción Genética
6.
Mol Biol Cell ; 12(2): 279-95, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11179415

RESUMEN

The cell adhesion molecule E-cadherin has been implicated in maintaining the polarized phenotype of epithelial cells and suppression of invasiveness and motility of carcinoma cells. Na,K-ATPase, consisting of an alpha- and beta-subunit, maintains the sodium gradient across the plasma membrane. A functional relationship between E-cadherin and Na,K-ATPase has not previously been described. We present evidence that the Na,K-ATPase plays a crucial role in E-cadherin-mediated development of epithelial polarity, and suppression of invasiveness and motility of carcinoma cells. Moloney sarcoma virus-transformed Madin-Darby canine kidney cells (MSV-MDCK) have highly reduced levels of E-cadherin and beta(1)-subunit of Na,K-ATPase. Forced expression of E-cadherin in MSV-MDCK cells did not reestablish epithelial polarity or inhibit the invasiveness and motility of these cells. In contrast, expression of E-cadherin and Na,K-ATPase beta(1)-subunit induced epithelial polarization, including the formation of tight junctions and desmosomes, abolished invasiveness, and reduced cell motility in MSV-MDCK cells. Our results suggest that E-cadherin-mediated cell-cell adhesion requires the Na,K-ATPase beta-subunit's function to induce epithelial polarization and suppress invasiveness and motility of carcinoma cells. Involvement of the beta(1)-subunit of Na,K-ATPase in the polarized phenotype of epithelial cells reveals a novel link between the structural organization and vectorial ion transport function of epithelial cells.


Asunto(s)
Movimiento Celular/fisiología , Polaridad Celular/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Actinas/metabolismo , Actinas/ultraestructura , Animales , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular/fisiología , Línea Celular/virología , Células Clonales , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Perros , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibroblastos , Riñón/citología , Riñón/metabolismo , Riñón/patología , Virus del Sarcoma Murino de Moloney , Subunidades de Proteína , Sodio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética
7.
Cancer Res ; 52(22): 6229-36, 1992 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-1423266

RESUMEN

Effective vaccination against cancer, either for prophylaxis or therapy, has been an elusive goal for years. Cytokine gene therapy offers a novel approach to generate immunogenic tumor cell vaccines. To examine the feasibility of cytokine gene transfer into human renal cancer (RC) cells, we introduced the cDNAs for human interleukin-2 (IL-2) or interferon-gamma (IFN-gamma) into various RC cell lines with retroviral vectors. Using the NIH3T3 amplification assay, no replication competent retroviral particles were detectable in cell culture supernatants taken from gene-modified RC cell lines. Efficient expression of both lymphokines was achieved. Depending on the cell line and the vector construct used, lymphokine gene-modified human RC cell lines released 4 to 29 units/10(6) cells of IL-2, or up to 10 units/10(6) cells of IFN-gamma within 48 h. Fluorescence-activated cell sorter analysis of SK-RC-29 cells releasing IFN-gamma showed increased expression of major histocompatibility complex class I antigen, beta 2-microglobulin, and ICAM-1, as well as induction of major histocompatibility complex class II antigen expression [human leukocyte antigen(HLA)-DR, -DP], but no changes in these cell surface markers were observed with SK-RC-29 cells releasing IL-2. Following in vitro gamma-irradiation with 5,000 or 10,000 rad, growth of lymphokine gene-modified RC cells was abrogated, but their capability to release lymphokine and express lymphokine-induced antigenic determinants, such as HLA-DR, was retained. Tumor formation by the human RC cell line SK-RC-29 in BALB/c nude mice was not affected by IFN-gamma secretion, but was inhibited by in vivo release of IL-2 from s.c. injected tumor cells. These studies demonstrate the feasibility of retroviral mediated lymphokine-gene transfer into human RC cells and suggest a means for generating autologous or HLA-matched allogeneic tumor cell vaccines for the treatment of patients with renal cell carcinoma.


Asunto(s)
Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Linfocinas/genética , Retroviridae/genética , Animales , Antígenos de Neoplasias/análisis , Carcinoma de Células Renales/microbiología , Carcinoma de Células Renales/fisiopatología , Supervivencia Celular/efectos de la radiación , ADN/genética , Relación Dosis-Respuesta en la Radiación , Rayos gamma , Expresión Génica/genética , Vectores Genéticos/genética , Humanos , Interferón gamma/biosíntesis , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-2/biosíntesis , Interleucina-2/genética , Interleucina-2/metabolismo , Neoplasias Renales/microbiología , Neoplasias Renales/fisiopatología , Linfocinas/biosíntesis , Linfocinas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fenotipo , Retroviridae/fisiología , Transfección , Trasplante Heterólogo , Células Tumorales Cultivadas , Replicación Viral/fisiología
8.
Cancer Res ; 57(17): 3629-34, 1997 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-9288760

RESUMEN

Prostate-specific membrane antigen (PSMA), initially defined by monoclonal antibody (mAb) 7E11, is a now well-characterized type 2 integral membrane glycoprotein expressed in a highly restricted manner by prostate epithelial cells. 7E11 has been shown to bind an intracellular epitope of PSMA that, in viable cells, is not available for binding. Herein, we report the initial characterization of the first four reported IgG mAbs that bind the external domain of PSMA. Competitive binding studies indicate these antibodies define two distinct, noncompeting epitopes on the extracellular domain of PSMA. In contrast to 7E11, these mAbs bind to viable LNCaP cells in vitro. In addition, they show strong immunohistochemical reactivity to tissue sections of prostate epithelia, including prostate cancer. These mAbs were also strongly reactive with vascular endothelium within a wide variety of carcinomas (including lung, colon, breast, and others) but not with normal vascular endothelium. These antibodies should prove useful for in vivo targeting to prostate cancer, as well as to the vascular compartment of a wide variety of carcinomas.


Asunto(s)
Anticuerpos Monoclonales , Especificidad de Anticuerpos , Antígenos de Neoplasias/inmunología , Antígenos de Superficie/inmunología , Inmunoglobulina G , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Antígenos de Neoplasias/análisis , Antígenos de Superficie/análisis , Unión Competitiva , Western Blotting , Reacciones Cruzadas , Endotelio Vascular/inmunología , Técnica del Anticuerpo Fluorescente Indirecta , Glutamato Carboxipeptidasa II , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Especificidad de Órganos
9.
Cancer Res ; 50(17): 5531-6, 1990 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-2386958

RESUMEN

We have reviewed our laboratory's efforts to establish continuous human renal cancer cell lines. During the 16-year period of 1972 through 1987, 498 successive attempts resulted in establishment of 63 renal cancer cell lines. Of these lines, 46 were derived from primary kidney tumors and 17 from metastatic sites (lung, brain, bone, and lymph node). Forty-three of these lines have been characterized with regard to morphology, growth kinetics, anchorage-independent growth, tumorigenicity in athymic nude mice, and expression of kidney cell surface antigens. These results were compared with data from primary short term cultures of normal kidney epithelium. The overall success rate of establishing continuous renal cancer cell lines was 12.7%. In general, no significant difference in success was noted based on whether the specimen was derived from a primary or a metastatic lesion. However, all successfully established lines were derived from tumors exhibiting clinically "aggressive" behavior. All cell lines expressed proximal tubular cell differentiation antigens. Significant morphological heterogeneity was observed among normal kidney as well as kidney cancer cell lines in vitro. No significant difference in doubling time was found between cell lines of renal cancer and passage 1 cultures of normal kidney epithelium. Twenty-one of 30 (70%) lines assayed formed clones on soft agar and 26 of 33 (79%) lines grew in athymic mice. Among the 25 lines which were assayed for both soft agar growth and tumorigenicity in nude mice, this pair of phenotypic traits were concordant in 17 lines (60%). Four lines (16%) grew on agar but not in mice, while four other lines (16%) failed to grow in agar but were tumorigenic in mice.


Asunto(s)
Neoplasias Renales/patología , Riñón/citología , Animales , Anticuerpos Monoclonales , Antígenos de Superficie/análisis , División Celular , Línea Celular , Técnicas de Cultivo/métodos , Células Epiteliales , Humanos , Cinética , Ratones , Ratones Desnudos , Trasplante Heterólogo
10.
Cancer Res ; 50(14): 4190-4, 1990 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-2364374

RESUMEN

Human leukocyte alpha-interferon (IFN-alpha) has significant antitumor activity in advanced renal cell carcinoma (RC), with approximately 15% (range, 5 to 29%) of patients subjected to IFN-alpha therapy exhibiting a major objective response. We assayed 16 RC cell lines for intrinsic sensitivity to the growth-inhibitory effects of recombinant IFN-alpha. Similar to results observed in patients, cultured RCs could be divided into those that are inhibited by IFN-alpha and those that are not. In addition, the IFN-alpha-sensitive or -resistant phenotype of cultured RCs was correlated with surface expression of six unrelated kidney-associated differentiation antigens. The expression of one antigen, a Mr 160,000 glycoprotein (gp160), was found to correlate with resistance to IFN-alpha. Proliferation of seven RC cell lines expressing gp160 (gp160+) was not significantly inhibited by IFN-alpha at concentrations as high as 3000 units/ml. In contrast, proliferation of eight of nine RC cell lines lacking expression of gp160 (gp160-) was markedly inhibited by IFN-alpha. The effect of IFN-alpha on gp160+ and gp160- RC xenografts in nu/nu mice was examined. In separate experiments, two gp160+ RC cell lines and five gp160- RC cell lines were injected s.c. into nu/nu mice; one half of the mice were subsequently treated with 10(6) units of IFN-alpha i.p. 3 times a wk, and one half received no IFN-alpha. Tumors appeared at the sites of inoculation in all mice given injections of gp160+ RC cell lines within 10 to 25 days regardless of INF-alpha therapy. Mice given injections of gp160- RC cell lines, but not receiving IFN-alpha, also formed tumors. In contrast, gp160- RC cell lines injected into mice that were treated with IFN-alpha exhibited a marked sensitivity, as demonstrated by either no tumor formation or delayed tumor formation. We conclude that the absence of gp160 expression by RCs may be predictive of sensitivity to the antitumor effects of IFN-alpha and, thus, provide a basis for identifying IFN-responsive patients.


Asunto(s)
Antígenos de Diferenciación/análisis , Carcinoma de Células Renales/tratamiento farmacológico , Glicoproteínas/análisis , Interferón Tipo I/farmacología , Neoplasias Renales/tratamiento farmacológico , Células Tumorales Cultivadas/citología , Animales , Anticuerpos Monoclonales , Antígenos de Diferenciación/biosíntesis , División Celular/efectos de los fármacos , Línea Celular , Femenino , Glicoproteínas/biosíntesis , Humanos , Interferón Tipo I/uso terapéutico , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Ratones , Ratones Desnudos , Proteínas Recombinantes , Trasplante Heterólogo , Células Tumorales Cultivadas/efectos de los fármacos
11.
Cancer Res ; 49(23): 6774-80, 1989 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-2684399

RESUMEN

Murine monoclonal antibody (mAb) F31 detects a heat-stable antigen (URO-8) found in the acidic lipid fraction of renal cancer cell extracts. Serological analysis of mAb F31 reactivity was assayed on 176 human cell lines. mAb F31 reacted with 38 of 45 renal cancers, a subpopulation of cells in primary cultures of normal renal epithelia, and two of 13 colon, two of 15 lung, and four of five ovarian cancers. No other epithelial, hematopoietic, or neuroectodermal cell lines tested were reactive. Immunofluorescence and immunoperoxidase analyses of fresh frozen tissue sections revealed mAb F31 reactivity in kidney, gastrointestinal tract, biliary canaliculi, bronchial epithelium, and skin. Within the kidney, mAb F31 immunoreactivity was confined to the straight portion of the proximal tubule. A panel composed of previously characterized mAbs as well as mAb F31 defines the antigenic phenotype of proximal convoluted tubular cells as URO-2+/URO-3+/URO-4+/URO-10+/URO-8-/URO-5-; proximal straight tubular cells as URO-2+/URO-3+/URO-4+/URO-10-/URO-8+/URO-5-; and cells of the descending thin limb of Henle as URO-2-/URO-3+ or -/URO-4+/URO-10-/URO-8-/URO-5+. While adult proximal tubular cells demonstrated reciprocal expression of URO-8 and URO-10, fetal kidney proximal tubule progenitor cells coexpressed both antigens (URO-10+/URO-8+). Fifty renal cancer specimens were typed with these antibodies. Fourteen cases were URO-10+/URO-8-, ten cases were URO-10-/URO-8+, and 25 cases expressed both antigens (URO-10+/URO-8+). These phenotypes are consistent with derivation of these particular subsets from the proximal convoluted tubule, the pars recta, or a proximal tubule progenitor cell, respectively. Only one specimen failed to express either URO-8 or URO-10.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/inmunología , Carcinoma de Células Renales/inmunología , Neoplasias Renales/inmunología , Túbulos Renales Proximales/inmunología , Anticuerpos Antineoplásicos/inmunología , Biomarcadores de Tumor , Diferenciación Celular , Técnica del Anticuerpo Fluorescente , Glucolípidos/inmunología , Humanos , Técnicas para Inmunoenzimas
12.
Cancer Res ; 57(12): 2362-5, 1997 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-9192809

RESUMEN

MN is a novel cell surface antigen originally detected in human HeLa cells. Although it is also expressed in normal gastric mucosa, this antigen was previously found to be expressed in cells with a malignant phenotype in certain tissues of the female genital tract (cervix and ovary). Using an oligonucleotide primer set specific for MN-complimentary DNA, we performed reverse transcription-PCR assays on RNAs extracted from human cell lines and tissues to evaluate whether this marker might be expressed at other sites. RNA libraries extracted from normal human heart, lung, kidney, prostate, peripheral blood, brain, placenta, and muscle were negative for MN expression. RNAs extracted from liver and pancreatic tissue were positive for MN expression. Three of six renal cancer cell lines tested revealed MN expression. In addition, 12 of 17 samples of human renal cell carcinoma tissue tested positive for MN, all 12 of which were clear cell adenocarcinomas. This survey identified a unique association of MN expression with renal cell cancers, especially those of the clear cell variety, suggesting that MN is a potential marker for the diagnosis, staging, and therapeutic monitoring of renal cell carcinoma in humans.


Asunto(s)
Antígenos de Neoplasias , Biomarcadores de Tumor/metabolismo , Anhidrasas Carbónicas , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Anhidrasa Carbónica IX , Carcinoma de Células Renales/diagnóstico , Femenino , Células HeLa/metabolismo , Humanos , Neoplasias Renales/diagnóstico , Masculino , Neoplasias de la Próstata/metabolismo , Distribución Tisular , Células Tumorales Cultivadas/metabolismo
13.
Cancer Res ; 60(18): 5237-43, 2000 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11016653

RESUMEN

Prostate-specific membrane antigen (PSMA) is a well-characterized cell surface antigen expressed by virtually all prostate cancers (PCas). PSMA has been successfully targeted in vivo with (111)In-labeled 7E11 monoclonal antibody (mAb; ProstaScint; Cytogen, Princeton, NJ), which binds to an intracellular epitope of PSMA. This work reports the in vitro characterization of three recently developed mAbs that bind the extracellular domain of PSMA (PSMAext). Murine mAbs J415, J533, J591, and 7E11 were radiolabeled with 131I and evaluated in competitive and saturation binding studies with substrates derived from LNCaP cells. J415 and J591 were conjugated to 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid labeled with (111)In. The uptake and cellular processing of these antibodies were evaluated in viable LNCaP cells. All four mAbs could be labeled with 131I up to a specific activity of 350 MBq/mg with no or little apparent loss of immunoreactivity. Competition assays revealed that J415 and J591 compete for binding to PSMAext antigen. J533 bound to a region close to the J591 binding epitope, but J533 did not interfere with J415 binding to PSMA. mAb 7E11 did not inhibit the binding of J415, J533, or J591 (or vice versa), consistent with earlier work that these latter mAbs bind PSMAext whereas 7E11 binds the intracellular domain of PSMA. Saturation binding studies demonstrated that J415 and J591 bound with a similar affinity (Kds 1.76 and 1.83 nM), whereas J533 had a lower affinity (Kd, 18 nM). In parallel studies, all four mAbs bound to a similar number of PSMA sites expressed by permeabilized cells (1,000,000-1,300,000 sites/cell). In parallel studies performed with viable LNCaP cells, J415, J533, and J591 bound to a similar number of PSMA sites (i.e., 600,000-800,000 sites/cell), whereas 7E11 bound only to a subpopulation of the available PSMA sites (95,000 sites/cell). This apparent binding of 7E11 to viable cells can be accounted for by a 5-7% subpopulation of permeabilized cells produced when the cells were trypsinized and suspended. Up to five DOTA chelates could be bound to either J415 or J591 without compromising immunoreactivity. A comparison of the cellular uptake and metabolic processing of the 131I- and (111)In-labeled antibodies showed a rapid elimination of 131I from the cell and a high retention of (111)In. All four mAbs recognized and bound to similar numbers of PSMAs expressed by ruptured LNCaP cells (i.e., the exposed intracellular and extracellular domains of PSMA). By comparison to J415 and J591, J533 had a lower binding affinity. Both J415 and J591 recognized and bound to the same high number of PSMAs expressed by intact LNCaP. By contrast, 7E11 bound to fewer sites expressed by intact LNCaP cells (i.e., the exposed extracellular domain of PSMA). Both J415 and J591 are promising mAbs for the targeting of viable PSMA-expressing tissue with diagnostic and therapeutic metallic radionuclides.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/inmunología , Antígenos de Superficie/inmunología , Carboxipeptidasas/inmunología , Inmunoconjugados/inmunología , Radioisótopos de Yodo , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacocinética , Especificidad de Anticuerpos , Antígenos de Neoplasias/metabolismo , Antígenos de Superficie/metabolismo , Unión Competitiva , Carboxipeptidasas/metabolismo , Membrana Celular/metabolismo , Quelantes/farmacocinética , Estabilidad de Medicamentos , Glutamato Carboxipeptidasa II , Humanos , Inmunoconjugados/metabolismo , Inmunoconjugados/farmacocinética , Radioisótopos de Indio , Radioisótopos de Yodo/uso terapéutico , Marcaje Isotópico , Cinética , Masculino , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Estructura Terciaria de Proteína , Control de Calidad , Células Tumorales Cultivadas
14.
Cancer Res ; 59(13): 3192-8, 1999 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10397265

RESUMEN

Prostate-specific membrane antigen (PSMA) is a type II integral membrane glycoprotein that was initially characterized by the monoclonal antibody (mAb) 7E11. PSMA is highly expressed in prostate secretory-acinar epithelium and prostate cancer as well as in several extraprostatic tissues. Recent evidence suggests that PSMA is also expressed in tumor-associated neovasculature. We examined the immunohistochemical characteristics of 7E11 and those of four recently developed anti-PSMA mAbs (J591, J415, and Hybritech PEQ226.5 and PM2J004.5), each of which binds a distinct epitope of PSMA. Using the streptavidin-biotin method, we evaluated these mAbs in viable prostate cancer cell lines and various fresh-frozen benign and malignant tissue specimens. In the latter, we compared the localization of the anti-PSMA mAbs to that of the anti-endothelial cell mAb CD34. With rare exceptions, all five anti-PSMA mAbs reacted strongly with the neovasculature of a wide spectrum of malignant neoplasms: conventional (clear cell) renal carcinoma (11 of 11 cases), transitional cell carcinoma of the urinary bladder (6 of 6 cases), testicular embryonal carcinoma (1 of 1 case), colonic adenocarcinoma (5 of 5 cases), neuroendocrine carcinoma (5 of 5 cases), glioblastoma multiforme (1 of 1 cases), malignant melanoma (5 of 5 cases), pancreatic duct carcinoma (4 of 4 cases), non-small cell lung carcinoma (5 of 5 cases), soft tissue sarcoma (5 of 6 cases), breast carcinoma (5 of 6 cases), and prostatic adenocarcinoma (2 of 12 cases). Localization of the anti-PSMA mAbs to tumor-associated neovasculature was confirmed by CD34 immunohistochemistry in sequential tissue sections. Normal vascular endothelium in non-cancer-bearing tissue was consistently PSMA negative. The anti-PSMA mAbs reacted with the neoplastic cells of prostatic adenocarcinoma (12 of 12 cases) but not with the neoplastic cells of any other tumor type, including those of benign and malignant vascular tumors (0 of 3 hemangiomas, 0 of 1 hemangioendothelioma, and 0 of 1 angiosarcoma). The mAbs to the extracellular PSMA domain (J591, J415, and Hybritech PEQ226.5) bound viable prostate cancer cells (LNCaP and PC3-PIP), whereas the mAbs to the intracellular domain (7E11 and Hybritech PM2J004.5) did not. All five anti-PSMA mAbs reacted with fresh-frozen benign prostate secretory-acinar epithelium (28 of 28 cases), duodenal columnar (brush border) epithelium (11 of 11 cases), proximal renal tubular epithelium (5 of 5 cases), colonic ganglion cells (1 of 12 cases), and benign breast epithelium (8 of 8 cases). A subset of skeletal muscle cells was positive with 7E11 (7 of 7 cases) and negative with the other four anti-PSMA mAbs. PSMA was consistently expressed in the neovasculature of a wide variety of malignant neoplasms and may be an effective target for mAb-based antineovasculature therapy.


Asunto(s)
Antígenos de Superficie , Carboxipeptidasas/análisis , Carboxipeptidasas/genética , Neoplasias/irrigación sanguínea , Neoplasias/enzimología , Neovascularización Patológica/enzimología , Próstata/enzimología , Anticuerpos Monoclonales , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/genética , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Carboxipeptidasas/inmunología , Carcinoma de Células Renales/irrigación sanguínea , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/patología , Femenino , Glutamato Carboxipeptidasa II , Humanos , Masculino , Neoplasias/patología , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Neoplasias Testiculares/irrigación sanguínea , Neoplasias Testiculares/enzimología , Neoplasias Testiculares/patología , Transfección , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/irrigación sanguínea , Neoplasias de la Vejiga Urinaria/enzimología , Neoplasias de la Vejiga Urinaria/patología
15.
Cancer Res ; 58(18): 4055-60, 1998 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9751609

RESUMEN

Prostate-specific membrane antigen (PSMA) is a cell surface glycoprotein expressed predominantly by prostate cancer cells. We have characterized four monoclonal antibodies that bind to the extracellular domain of PSMA (Liu et al., Cancer Res., 57: 3629-3634, 1997). Here we report that viable LNCaP cells internalize these antibodies. Laser scanning confocal microscopy reveals that the internalized antibodies accumulate in endosomes, and immunoelectron microscopy reveals that endocytosis of the PSMA-antibody complex occurs via clathrin-coated pits. In addition, a quantitative cell surface biotinylation assay demonstrates that PSMA is constitutively endocytosed in LNCaP cells and that anti-PSMA antibodies increase the rate of internalization of PSMA. These studies suggest that PSMA might function as a receptor mediating the internalization of a putative ligand. The availability of prostate-specific internalizing antibodies should aid the development of novel therapeutic methods to target the delivery of toxins, drugs, or short-range isotopes specifically to the interior of prostate cancer cells.


Asunto(s)
Antígenos de Superficie/metabolismo , Carcinoma/metabolismo , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Antígenos de Superficie/inmunología , Endosomas/metabolismo , Humanos , Masculino , Microscopía Confocal , Antígeno Prostático Específico/inmunología , Células Tumorales Cultivadas
16.
Cancer Res ; 59(12): 2838-42, 1999 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-10383143

RESUMEN

The antitumor effect of immuno- and chemotherapeutic agents is executed through stimulation of apoptotic programs in susceptible cells. Apoptosis induced in tumor cells requires activation of members of the caspase family of proteases. Deficient expression or activation of caspases may account in part for the failure of many current anticancer therapies. However, recent studies suggest that cell death can proceed in the absence of caspases. We investigated the susceptibility of human renal cell carcinoma (RCC) lines to two distinct modes of cell death, apoptosis and necrosis. RCC lines displayed almost complete resistance to apoptosis in response to the intracellular zinc chelator, N,N,N'N'-tetrakis (2-pyridylmethyl) ethylenediamine (TPEN), which instead induced dramatic accumulation of nonapoptotic necrotic cells. Conversely, TPEN was a potent inducer of apoptosis in caspase-competent normal kidney cells (NK-72) and Jurkat T lymphocytes. Resistance to apoptosis in RCC lines correlated with almost complete loss of caspase-3 expression and variable down-regulation of caspase-7, caspase-8, and caspase-10. These data may explain the resistance of RCC to drugs inducing apoptosis and have important consequences for further attempts to manipulate tumor cell death.


Asunto(s)
Carcinoma de Células Renales/patología , Caspasas/metabolismo , Neoplasias Renales/patología , Apoptosis , Carcinoma de Células Renales/enzimología , Caspasas/deficiencia , Inhibidores de la Colinesterasa/farmacología , Etilenodiaminas/farmacología , Humanos , Células Jurkat , Neoplasias Renales/enzimología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/patología , Necrosis , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Células Tumorales Cultivadas
17.
Cancer Res ; 61(5): 2008-14, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11280760

RESUMEN

A theoretical drawback to alpha-particle therapy with 213Bi is the short range of the particle track coupled with the short half-life of the radionuclide, thereby potentially limiting effective cytotoxicity to rapidly accessible, disseminated individual tumor cells (e.g., as in leukemia). In this work, a prostate carcinoma spheroid model was used to evaluate the feasibility of targeting micrometastatic clusters of tumor cells using 213Bi-labeled anti-prostate-specific membrane antigen (PSMA) antibody, J591. In prostate cancer, vascular dissemination of tumor cells or tumor cell clusters to the marrow constitutes an important step in the progression of this disease to widespread skeletal involvement, an incurable state. Such prevascularized clusters are ideal targets for radiolabeled antibodies because the barriers to antibody penetration that are associated with the capillary basal lamina have not yet formed. Beta- and gamma-emitting radionuclides such as 131I, which are widely used in radioimmunotherapy, are not expected to be effective when targeting single cells or small cell clusters. This is because the range of the emissions is one to two orders of magnitude greater than the target size, and the energy deposited per traversal is insufficient to produce any significant radiobiological effect. Spheroids of the prostate cancer cell line, LNCaP-LN3, were used as a model of prevascularized micrometastases; their response to an anti-PSMA antibody, J591, radiolabeled with the alpha-particle emitter 213Bi (T(1/2), 45.6 min.) has been measured. The time course of spheroid volume reductions was found to be sensitive to the initial spheroid volume. J591 labeled with 0.9 MBq/ml 213Bi resulted in a 3-log reduction in spheroid volume on day 33, relative to control, for spheroids with an initial diameter of 130 microm; 1.8 MBq/ml were required to achieve a similar response for spheroids with an initial diameter of 180 microm. Equivalent spheroid responses were observed after 12 Gy of acute external beam photon irradiation. Monte Carlo-based microdosimetric analyses of the 213Bi decay distribution in individual spheroids of 130-microm diameter yielded an average alpha-particle dose of 3.7 Gy to the spheroids, resulting in a relative biological effectiveness factor of 3.2 over photon irradiation. The activity concentrations used in the experiments were clinically relevant, and this work supports the possibility of using 213Bi-labeled antibodies not only for disseminated single tumor cells, as found in patients with leukemia, but also for micrometastatic tumor deposits up to 180 microm in diameter (1200 cells).


Asunto(s)
Antígenos de Superficie , Bismuto/farmacología , Carboxipeptidasas/inmunología , Inmunotoxinas/farmacología , Neoplasias de la Próstata/radioterapia , Radioisótopos/farmacología , Partículas alfa/uso terapéutico , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , División Celular/efectos de la radiación , Glutamato Carboxipeptidasa II , Humanos , Inmunotoxinas/inmunología , Inmunotoxinas/metabolismo , Masculino , Microscopía Confocal , Metástasis de la Neoplasia , Neoplasias de la Próstata/inmunología , Radioinmunoterapia , Esferoides Celulares/inmunología , Esferoides Celulares/efectos de la radiación , Células Tumorales Cultivadas
18.
Cancer Res ; 60(21): 6095-100, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11085533

RESUMEN

A novel alpha-particle emitting monoclonal antibody construct targeting the external domain of prostate-specific membrane antigen (PSMA) was prepared and evaluated in vitro and in vivo. The chelating agent, N-[2-amino-3-(p-isothiocyanatophen-yl)propyl]-trans-cyclohexane-1, 2-diamine-N,N',N',N'',N''-pentaacetic acid, was appended to J591 monoclonal antibody to stably bind the 213Bi radiometal ion. Bismuth-213 is a short-lived (t 1/2 = 46 min) radionuclide that emits high energy alpha-particles with an effective range of 0.07-0.10 mm that are ideally suited to treating single-celled neoplasms and micrometastatic carcinomas. The LNCaP prostate cancer cell line had an estimated 180,000 molecules of PSMA per cell; J591 bound to PSMA with a 3-nM affinity. After binding, the radiolabeled construct-antigen complex was rapidly internalized into the cell, carrying the radiometal inside. [213Bi]J591 was specifically cytotoxic to LNCaP. The LD50 value of [213Bi]J591 was 220 nCi/ml at a specific activity of 6.4 Ci/g. The potency and specificity of [213Bi]J591 directed against LNCaP spheroids, an in vitro model for micrometastatic cancer, also was investigated. [213Bi]J591 effectively stopped growth of LNCaP spheroids relative to an equivalent dose of the irrelevant control [213Bi]HuM195 or unlabeled J591. Cytotoxicity experiments in vivo were carried out in an athymic nude mouse model with an i.m. xenograft of LNCaP cells. [213Bi]J591 was able to significantly improve (P < 0.0031) median tumor-free survival (54 days) in these experiments relative to treatment with irrelevant control [213Bi]HuM195 (33 days), or no treatment (31 days). Prostate-specific antigen (PSA) was also specifically reduced in treated animals. At day 51, mean PSA values were 104 ng/ml +/- 54 ng/ml (n = 4, untreated animals), 66 ng/ml +/- 16 ng/ml (n = 6, animals treated with [213Bi]HuM195), and 28 ng/ml +/- 22 ng/ml (n = 6, animals treated with [213Bi]J591). The reduction of PSA levels in mice treated with [213Bi]J591 relative to mice treated with [213Bi]HuM195 and untreated control animals was significant with P < 0.007 and P < 0.0136, respectively. In conclusion, a novel [213Bi]-radiolabeled J591 has been constructed that selectively delivers alpha-particles to prostate cancer cells for potent and specific killing in vitro and in vivo.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Bismuto/farmacología , Inmunotoxinas/farmacología , Neoplasias de la Próstata/radioterapia , Radioinmunoterapia , Radioisótopos/farmacología , Partículas alfa/uso terapéutico , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Complejo Antígeno-Anticuerpo/metabolismo , Sitios de Unión , Muerte Celular/efectos de la radiación , Humanos , Inmunotoxinas/inmunología , Inmunotoxinas/metabolismo , Cinética , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Esferoides Celulares/efectos de la radiación , Especificidad por Sustrato , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Clin Oncol ; 7(12): 1885-91, 1989 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2555451

RESUMEN

Forty patients with metastatic, recurrent, or unresectable renal cell carcinoma were entered into a study of the therapeutic efficacy of adoptive immunotherapy using periodate (IO4-) and interleukin-2 (IL2)-activated autologous leukocytes and continuous infusion low-dose IL2. Patient survival was also examined. The first 15 consecutive patients were enrolled in protocol A without an IL2 priming phase and the following 25 patients were entered in protocol B where a 5-day priming phase was initiated before leukapheresis. A maintenance regimen consisted of either 3 x 10(6) units of recombinant interferon-alpha (rIFN-alpha), three times per week only or together with leukapheresis and infusion of IO4-/IL2-activated cells and 2 days of continuous infusion IL2 per month. Thirty-four patients completed the protocol treatment. Four patients were removed from the study owing to rapid tumor progression and two patients died while receiving treatment. The clinical response rate was 22%: two patients had a complete response and five patients had a partial response. Among the 25 patients who had no clinical response, 11 patients had either a mixed response or stabilization. Neither response, response duration, nor site response correlated with the total dose of IL2 administered or the number of activated killer cells infused. Patients who received maintenance therapy had longer survival times than patients who did not receive such therapy. All toxicity and side effects associated with IL2 treatment were transient and resolved after discontinuation of the drug. Patients on maintenance therapy tolerated both rIFN-alpha and monthly infusions of activated killer cells and IL2 well. This study confirms the concept of adoptive immunotherapy as a new treatment approach for advanced renal cell carcinoma and suggests that maintenance therapy may prolong survival time.


Asunto(s)
Carcinoma de Células Renales/terapia , Interleucina-2/administración & dosificación , Neoplasias Renales/terapia , Células Asesinas Activadas por Linfocinas/inmunología , Adulto , Anciano , Esquema de Medicación , Evaluación de Medicamentos , Humanos , Inmunización Pasiva , Inmunoterapia/efectos adversos , Interferón Tipo I/uso terapéutico , Activación de Linfocitos/efectos de los fármacos , Persona de Mediana Edad , Nefrectomía , Ácido Peryódico/farmacología , Proteínas Recombinantes
20.
J Clin Oncol ; 11(4): 738-50, 1993 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8478666

RESUMEN

PURPOSE: To define the imaging and biodistribution characteristics of iodine 131-labeled monoclonal antibody (mAb) G250 (131I-mAbG250), which recognizes a cell-surface antigen expressed by human renal cell carcinoma (RCC). PATIENTS AND METHODS: G250 is a cell-surface antigen recognized by mAbG250 expressed by RCC but not detected in normal kidney. Clear-cell RCC, the most frequent form of RCC, shows homogeneous expression of G250, whereas non-clear-cell RCC and cancers derived from other organs generally do not express G250. Expression in normal tissues is highly restricted and limited to large bile ducts and gastric epithelium. 131I-mAbG250 was administered intravenously (IV) to 16 patients with RCC 7 to 8 days before surgery at five dose levels, with at least three patients entered at each dose level. RESULTS: Clear tumor images were observed in 12 patients with G250-positive tumors and in one of three patients with G250-negative tumors. Imaged lesions in the peritoneal cavity were confirmed at surgery. The smallest lesion visualized was 8 mm in diameter. The specificity of 131I-mAbG250 localization to tumor tissue was established by radioactivity measurements, autoradiography, and immunohistochemistry of biopsied tissues, and technetium 99-human serum albumin blood-flow studies. The fraction of the injected 131I-mAbG250 dose per gram tumor (%ID/g tumor) localized in G250-positive tumors showed a broad range, but reached levels as high as 0.02% to 0.12%. CONCLUSION: 131I-mAbG250 localized specifically to G250 antigen-positive RCC and seems to have considerable potential as an imaging agent in RCC patients. 131I-mAbG250 uptake in the tumors, relative as well as absolute, are among the highest reported for tumor biopsies obtained 8 days after IV mAb administration. Based on the specific localization and high accumulation, mAb G250 may have therapeutic potential.


Asunto(s)
Carcinoma de Células Renales/diagnóstico por imagen , Neoplasias Renales/diagnóstico por imagen , Radioinmunodetección , Adulto , Anciano , Animales , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/inmunología , Autorradiografía , Femenino , Humanos , Inmunohistoquímica , Técnicas In Vitro , Radioisótopos de Yodo/análisis , Radioisótopos de Yodo/farmacocinética , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Radioinmunodetección/efectos adversos , Agregado de Albúmina Marcado con Tecnecio Tc 99m
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA