Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Med Oncol ; 29(3): 1486-94, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21769502

RESUMO

Cell surface transmembrane signaling receptors EGFR, HER3, and HER4 are activated by ligand-binding-mediated dimerization and phosphorylation. In contrast, HER2 amplification promotes signaling by increasing homo/heterodimerization and ligand binding. Trastuzumab or lapatinib therapy of HER2 amplicon-positive breast cancer cells induces growth inhibition and intracellular growth pathway signaling modulation. The mechanism(s) by which trastuzumab, an IgG1 humanized antibody, induces modification of cell signaling upon binding to an extracellular determinant on a ligand-less "receptor" membrane protein remains unexplained. Using immune detection methodology comprised of antibodies detecting three distinct domains of HER and five tyrosine/threonine phosphorylation sites, the effects of trastuzumab and lapatinib were defined during steady state growth inhibition. Here, we show that lapatinib markedly reduces HER2 tyrosine phosphorylation, while in contrast, no change in tyrosine phosphate levels is detected during trastuzumab-mediated cell growth inhibition. As trastuzumab treatment does not change either the steady state HER2 protein levels or HER2 mRNA, these findings argue against an antibody-dependent alteration in internalization kinetics. We further show a sequenced relationship between lapatinib-induced blockage of phosphorylation (6-8 h) and induction of delayed cell death (5-6 days), while trastuzumab-treated cells showed no evidence of cell death up to 9 days. Taken together, these results demonstrate that inhibition of HER2 phosphorylation by lapatinib is sufficient to induce apoptosis while trastuzumab binding to the extracellular HER2 domain may function by sterically modulating the detection of phosphate moieties by cytoplasmic signal transducers. This investigation also detected a 20 kD protein, which is down-regulated by lapatinib, further demonstrating the complexity of this signal transduction system.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Western Blotting , Feminino , Humanos , Lapatinib , Fosforilação , Receptor ErbB-2/efeitos dos fármacos , Receptor ErbB-2/metabolismo , Treonina/metabolismo , Trastuzumab , Tirosina/metabolismo
2.
Br J Cancer ; 88(6): 937-9, 2003 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-12644833

RESUMO

A recombinant fusion protein of colon carcinoma binding A33 single chain antibody with cytosine deaminase displayed specific antigen binding and enzyme activity in surface plasmon resonance and is catalytic activity assay. In vitro, it selectively increased the toxicity of 5-FC to A33 antigen-positive cells by 300-fold, demonstrating the potency of this ADEPT strategy.


Assuntos
Glicoproteínas de Membrana/imunologia , Nucleosídeo Desaminases/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Antígenos de Neoplasias , Catálise , Citosina Desaminase , Escherichia coli/genética , Humanos , Fragmentos de Imunoglobulinas , Região Variável de Imunoglobulina , Glicoproteínas de Membrana/farmacologia , Nucleosídeo Desaminases/química , Nucleosídeo Desaminases/imunologia , Reação em Cadeia da Polimerase , Células Tumorais Cultivadas
3.
Cancer Res ; 61(18): 6851-9, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11559561

RESUMO

Mouse monoclonal antibody A33 (mAb A33) recognizes a M(r) 43,000 cell surface glycoprotein (designated A33) expressed in human colonic epithelium and colon cancer but absent from most other normal tissues. In patients, mAb A33 localizes with high specificity to colon cancer and is retained for up to 6 weeks in the cancer but cleared rapidly from normal colon (5-6 days). As a carrier of (125)I or (131)I, mAb A33 has shown antitumor activity. Induction of strong human anti-mouse antibody (immunoglobulin; HAMA) responses in patients, however, limits the use of the murine mAb A33 to very few injections. A humanized version of this antibody (huAb A33) has been prepared for Phase I and II clinical studies in patients with colon cancer. In those studies, immunogenicity of huAb A33 has been monitored using a novel, highly sensitive BIACORE method, which allows measurement of human anti-human antibodies (HAHAs) without the use of secondary reagents. We found that 63% (26 of 41) of the patients treated with repeated doses of huAb A33 developed HAHAs against a conformational antigenic determinant located in the V(L) and V(H) regions of huAb A33. Detailed serological analysis showed two distinct types of HAHAs. HAHA of type I (49% of patients) was characterized by an early onset with peak HAHA levels after 2 weeks of treatment, which declined with ongoing huAb A33 treatment. HAHA of type II (17% of patients) was characterized by a typically later onset of HAHA than in type I and by progressively increasing HAHA levels with each subsequent huAb A33 administration. Colon cancer patients with type I HAHAs did not develop infusion-related adverse events. In contrast, HAHA of type II was indicative of infusion-related adverse events. By using this new method, we were able to distinguish these two types of HAHAs in patients while on antibody treatment, allowing patients to be removed from study prior to the onset of severe infusion-related adverse events.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/sangue , Neoplasias do Colo/imunologia , Glicoproteínas de Membrana/imunologia , Sequência de Aminoácidos , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/imunologia , Anticorpos Antineoplásicos/biossíntese , Anticorpos Antineoplásicos/imunologia , Antígenos de Neoplasias/imunologia , Técnicas Biossensoriais , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Neoplasias do Colo/terapia , Mapeamento de Epitopos , Epitopos/imunologia , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/sangue , Imunoglobulina M/biossíntese , Imunoglobulina M/sangue , Dados de Sequência Molecular , Fragmentos de Peptídeos/imunologia
4.
J Nucl Med ; 42(8): 1251-6, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11483688

RESUMO

UNLABELLED: A33, a monoclonal antibody that targets colon carcinomas, was labeled with (125)I or (131)I and the relative therapeutic efficacy of the 2 radiolabeled species was compared in a human colon cancer xenograft system. METHODS: Nude mice bearing human SW1222 colon carcinoma xenografts were administered escalating activities of (125)I-A33 (9.25-148 MBq) or (131)I-A33 (0.925-18.5 MBq), (125)I- and (131)I-labeled control antibodies, unlabeled antibody, or no antibody. The effects of treatment were assessed using the endpoints of tumor growth delay and cure. RESULTS: Tumor growth delay increased with administered activity for all radiolabeled antibodies. Approximately 4.5 times more activity was required for (125)I-A33 to produce therapeutic effects that were equivalent to those of (131)I-A33. This ratio was approximately 7 for a nonspecific, noninternalizing isotype-matched, radiolabeled control antibody. Unlabeled A33 antibody had no effect on tumor growth. Approximately 10 times more activity of (125)I-A33 produced toxicity similar to that of (131)I-A33, and this ratio fell to approximately 6 for radiolabeled control antibody. CONCLUSION: Treatment with (125)I-A33 resulted in a relative therapeutic gain of approximately 2 compared with (131)I-A33 in this experimental system.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias do Colo/radioterapia , Glicoproteínas de Membrana/imunologia , Radioimunoterapia , Compostos Radiofarmacêuticos/uso terapêutico , Animais , Neoplasias do Colo/patologia , Feminino , Humanos , Radioisótopos do Iodo , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transplante Heterólogo
5.
J Nucl Med ; 42(8): 1281-7, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11483692

RESUMO

UNLABELLED: Absorbed doses in (90)Y radioimmunotherapy are usually estimated by extrapolating from (111)In imaging data. PET using (86)Y (beta(+) 33%; half-life, 14.7 h) as a surrogate radiolabel could be a more accurate alternative. The aim of this study was to evaluate an (86)Y-labeled monoclonal antibody (mAb) as a PET imaging agent and to compare the biodistribution of (86)Y- and (111)In-labeled mAb. METHODS: The humanized anti-Lewis Y mAb hu3S193 was labeled with (111)In or (86)Y through CHX-A"-diethylenetriaminepentaacetic acid chelation. In vitro cell binding and cellular retention of radiolabeled hu3S193 were evaluated using HCT-15 colon carcinoma cells, a cell line expressing Lewis Y. Nude mice bearing HCT-15 xenografts were injected with (86)Y-hu3S193 or (111)In-hu3S193. The biodistribution was studied by measurements of dissected tissues as well as by PET and planar imaging. RESULTS: The overall radiochemical yield in hu3S193 labeling and purification was 42% +/- 2% (n = 2) and 76% +/- 3% (n = 6) for (86)Y and (111)In, respectively. Both radioimmunoconjugates specifically bound to HCT-15 cells. When cellular retention of hu3S193 was studied using (111)In-hu3S193, 80% of initially cell-bound (111)In activity was released into the medium as high-molecular-weight compounds within 8 h. When coadministered, in vivo tumor uptake of (86)Y-hu3S193 and (111)In-hu3S193 reached maximum values of 30 +/- 6 and 29 +/- 6 percentage injected dose per gram and tumor sites were easily identifiable by PET and planar imaging, respectively. CONCLUSION: At 2 d after injection of (111)In-hu3S193 and (86)Y-hu3S193 radioimmunoconjugates, the uptake of (111)In and (86)Y activity was generally similar in most tissues. After 4 d, however, the concentration of (86)Y activity was significantly higher in several tissues, including tumor and bone tissue. Accordingly, the quantitative information offered by PET, combined with the presumably identical biodistribution of (86)Y and (90)Y radiolabels, should enable more accurate absorbed dose estimates in (90)Y radioimmunotherapy.


Assuntos
Anticorpos Monoclonais , Compostos Radiofarmacêuticos , Tomografia Computadorizada de Emissão/métodos , Animais , Antígenos de Neoplasias/metabolismo , Autorradiografia , Neoplasias do Colo/diagnóstico por imagem , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Humanos , Imuno-Histoquímica , Radioisótopos de Índio , Camundongos , Camundongos Nus , Modelos Biológicos , Transplante de Neoplasias , Distribuição Tecidual , Transplante Heterólogo , Células Tumorais Cultivadas , Radioisótopos de Ítrio
6.
Cancer Res ; 61(11): 4474-82, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11389078

RESUMO

The chimeric monoclonal antibody KM871, directed against the G(D3) antigen, is under evaluation for its potential to target melanoma. To facilitate the in vivo evaluation of biodistribution properties and measurement of pharmacokinetics, KM871 was radiolabeled with (125)I via tyrosine residues and with (111)In via the bifunctional metal ion chelator C-functionalized trans-cyclohexyl diethylenetriaminepentaacetic acid (CHX-A"-DTPA) to lysine residues. Using antigen-positive SK-MEL-28 melanoma cells, immunoreactivities of 42 and 40% cell binding were obtained, respectively, for the two radioconjugates. Binding was enhanced in the presence of added unlabeled antibody. A humanized A33 antibody was similarly labeled with the two isotopes and used as a control. To determine and compare in vivo biodistribution characteristics of KM871 radiolabeled with (111)In or (125)I, mixtures of the radioconjugates were injected i.v. into BALB/c nude mice bearing G(D3)-positive-SK-MEL-28 melanoma xenografts. Gamma camera images were acquired; groups of five mice were sacrificed at various time intervals, and tumors, blood, and tissues were analyzed. (111)In-labeled CHX-A"-DTPA-KM871 showed a maximum tumor uptake of 41.9 +/- 7.0% injected dose/g at 72 h with prolonged retention over a 15-day period. The tumor:blood ratio was 3:1 by 72 h, and higher ratios were observed at later time points. No abnormal accumulation of (111)In-labeled conjugate was found in normal tissues. In contrast, there was little accumulation of (125)I-labeled KM871 in the same tumors. The specificity of antibody localization was confirmed by the low tumor uptake values for radiolabeled control antibody. Gamma camera imaging demonstrated excellent uptake of (111)In-labeled CHX-A"-DTPA-KM871 in the xenografts. Chromatographic analyses of xenograft cytosolic extracts demonstrated tumor internalization and catabolism of radiolabeled KM871 with the formation of small molecular weight metabolites. Laser scanning confocal microscopy demonstrated that the majority of intracellular KM871 is localized to lysosomes. Despite the catabolism of the radioconjugate, a dose-dependent increase in KM871 tumor localization was shown through immunohistochemical examination of xenograft biopsies. This study demonstrates for the first time the in vivo localization of a radiolabeled anti-G(D3) monoclonal antibody to G(D3)-expressing xenografts using gamma camera scanning techniques and tumor cell internalization of KM871 tagged with a green fluorescent dye, Alexa Fluor 488, through confocal microscopy. KM871 has potential for targeting tumors in patients with melanoma.


Assuntos
Anticorpos Monoclonais/farmacocinética , Gangliosídeos/imunologia , Imunoconjugados/farmacocinética , Melanoma/diagnóstico por imagem , Melanoma/metabolismo , Ácido Pentético/análogos & derivados , Compostos Radiofarmacêuticos/farmacocinética , Animais , Anticorpos Monoclonais/imunologia , Quelantes/química , Quelantes/farmacocinética , Feminino , Câmaras gama , Gangliosídeos/biossíntese , Humanos , Imuno-Histoquímica , Radioisótopos de Índio/química , Radioisótopos do Iodo/química , Isotiocianatos/química , Isotiocianatos/farmacocinética , Marcação por Isótopo , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ácido Pentético/química , Ácido Pentético/farmacocinética , Cintilografia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacocinética , Distribuição Tecidual , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Nucl Med ; 42(5): 764-9, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11337573

RESUMO

UNLABELLED: Radiolabeling monoclonal antibodies (mAbs) allows the evaluation of biodistribution of constructs in vivo through gamma camera imaging and also permits quantitation of mAb uptake in tumors through biopsy-based counting techniques. The quantitation of radiolabeled mAb uptake in cancer patients is complicated by the attenuation of gamma emissions of routinely used isotopes (e.g., 131I and 111In) and the spatial resolution and sensitivity of gamma cameras. METHODS: We used the positron-emitting isotope 124I (half-life [T1/2] = 4.2 d) to label the recombinant humanized anti-colorectal cancer A33 antibody (huA33) and evaluated its biodistribution properties and PET imaging characteristics in BALB/c nude mice bearing SW1222 colorectal xenografts and control colon tumors. RESULTS: The immunoreactivity of radioconjugate was 78% as determined using the cell-binding Lindmo assay. The apparent association constant was found to be 2.2 x 10(9) M(-1), and the number of antibody binding sites per cell was 371,000. The radioconjugate was found to be stable in serum obtained from mice at various times after injection. Assuming a two-compartment model with a four-parameter fit of mean blood levels, the T1/2alpha was 1.5 h and the T1/2beta was 38.2 h. Excellent tumor uptake was obtained, with maximal uptake reaching 50.0 +/- 7.0 percentage injected dose per gram of tumor by 4 d after injection. Specificity of localization was shown by lack of uptake in control tumor. PET imaging detected antigen-positive tumor by 4 h after injection, and high-resolution images were obtained by 24 h after injection. CONCLUSION: In clinical trials using PET, huA33 labeled with 124I has potential for imaging and staging colon tumors and quantifying antibody uptake in colon tumors in vivo.


Assuntos
Anticorpos Monoclonais , Neoplasias Colorretais/diagnóstico por imagem , Radioisótopos do Iodo , Glicoproteínas de Membrana/imunologia , Radioimunodetecção , Tomografia Computadorizada de Emissão , Animais , Anticorpos Monoclonais/farmacocinética , Antígenos de Neoplasias/imunologia , Neoplasias Colorretais/imunologia , Feminino , Humanos , Radioisótopos do Iodo/farmacocinética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacocinética , Distribuição Tecidual
8.
Br J Clin Pharmacol ; 51(2): 177-80, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11259992

RESUMO

AIMS: The population pharmacokinetics of 131I-mAbF19, a radiolabelled murine monoclonal antibody against fibroblast activation protein and a potential antitumour stroma agent, were investigated during two phase I studies in cancer patients. METHODS: 131I-mAbF19 serum concentration-time data were obtained in 16 patients from two studies involving imaging and dosimetry in colorectal carcinoma and soft tissue sarcoma. Doses of 0.2, 1 and 2 mg antibody were administered as 60 min intravenous infusions. The data were analysed by nonlinear mixed effect modelling. RESULTS: The data were described by a two-compartment model. Population mean values were 109 ml h(-1) for total serum clearance, 3.1 l for the volume of distribution of the central compartment, and 4.9 l for the volume of distribution at steady state. Mean terminal half-life was 38 h. Intersubject variability was high, but no patient covariates could be identified that further explained this variability. In particular, there was no influence of tumour type or mAbF19 dose. CONCLUSIONS: The pharmacokinetics of antistromal mAbF19 were well defined in these two studies with different solid tumour types, and were comparable with those of other murine monoclonal antibodies that do not bind to normal tissue antigens or blood cells.


Assuntos
Anticorpos Monoclonais/farmacocinética , Antígenos de Neoplasias , Antineoplásicos/farmacocinética , Biomarcadores Tumorais , Substâncias de Crescimento/imunologia , Neoplasias/metabolismo , Serina Endopeptidases/imunologia , Adulto , Idoso , Anticorpos Monoclonais/sangue , Antineoplásicos/sangue , Endopeptidases , Feminino , Gelatinases , Humanos , Masculino , Proteínas de Membrana , Pessoa de Meia-Idade
9.
J Nucl Med ; 41(11): 1905-12, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11079503

RESUMO

UNLABELLED: The purpose of this study was to determine the optimum sequence for combined modality therapy with radiolabeled antibodies and fractionated external beam radiation. METHODS: The uptake and distribution of a nontherapeutic activity of 125I-labeled tumor-associated A33 monoclonal antibody was determined in SW1222 human colon carcinoma xenografts in nude mice for 4 study groups: group 1, radiolabeled antibody alone; group 2, radiolabeled antibody administered (day 0) immediately before the first of 5 daily fractions of 2-Gy, 320-kilovolt peak x-rays; group 3, radiolabeled antibody administered after the fifth radiation fraction (day 5); and group 4, radiolabeled antibody administered 5 d after irradiation (day 10). Tumors were excised 5 d after antibody administration. Tumors were frozen and sectioned for histology and phosphor plate autoradiography. The percentage of A33 antigen-expressing cells was estimated by immunohistochemical staining. RESULTS: The average tumor uptake values relative to control group 1 were 1.47 (group 2), 0.78 (group 3), and 0.21 (group 4), which illustrates that tumor uptake is increased by almost 50% when the antibody is present in the blood at the start of irradiation. Five days into a fractionated irradiation protocol, antibody uptake was reduced, falling more significantly on day 10. Phosphor plate autoradiographs showed decreased uptake uniformity for groups 3 and 4. Immunohistochemical data showed a reduction in A33 antigen-positive cells from 85%, 64%, 50%, to 41% for groups 1-4, respectively. CONCLUSION: Maximum radiolabeled antibody tumor uptake was achieved when the antibody was administered just before radiation therapy. This might be explained by a transient increase in capillary leakage to macromolecules, followed by a reduction at later times, possibly the result of capillary damage and occlusion.


Assuntos
Neoplasias Colorretais/radioterapia , Radioimunoterapia , Animais , Anticorpos Monoclonais/imunologia , Antígenos/análise , Autorradiografia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Fracionamento da Dose de Radiação , Camundongos , Camundongos Nus , Transplante de Neoplasias , Dosagem Radioterapêutica , Células Tumorais Cultivadas
10.
Int J Cancer ; 87(3): 382-90, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10897044

RESUMO

Therapeutic proteins have been conjugated with polyethylene glycol (PEGylation) to reduce immunogenicity and enhance circulating dose. Here we have investigated the effect of PEGylation on immunogenicity, pharmacokinetics, and histologic microdistribution of tumor-targeting antibodies with humanized A33 antibody (huA33) as a model system. Conjugation of huA33 with methoxy-PEG of M(r) 5,000 (32%-34% of primary amines modified) or M(r) 20,000 (16%-18% modification) preserved >50% of native huA33 binding to SW1222 colon cancer cells. In mice, both PEGylated forms cleared from serum moderately slower than native huA33. After repeated immunization with PEG-huA33, antiantibody titers in immunocompetent mice were <5% of those in huA33-treated controls. Both PEG-huA33 forms reached approx. 75% of the maximum tumor dose of huA33 in SW1222-xenografted mice, but their tumor:blood ratios were considerably reduced. To demonstrate immunologic specificity of PEG-huA33 targeting in SW1222 tumor-bearing mice, antigenic sites were presaturated by injecting excess native huA33. This reduced subsequent uptake of PEG-huA33 by up to 80%, whereas presaturation with hu3S193 control antibody had no significant effect. To assess the microdistribution of antibody uptake in the same xenograft model, tumor tissue resected at different time points after antibody administration was examined for human IgG by immunohistochemistry. Both PEG preparations achieved the same peak staining intensity and homogeneity as native huA33 with a delay of several hours. Given the measured reduction in immunoreactivity in vitro, these results demonstrate that the tumor targeting potential of huA33 in vivo is preserved at PEGylation levels sufficient to suppress immunogenicity.


Assuntos
Adenocarcinoma/imunologia , Anticorpos Monoclonais/farmacocinética , Antígenos de Neoplasias/imunologia , Neoplasias do Colo/imunologia , Glicoproteínas de Membrana/imunologia , Polietilenoglicóis/farmacocinética , Animais , Anticorpos Monoclonais/análise , Anticorpos Monoclonais/imunologia , Feminino , Meia-Vida , Humanos , Imunização , Técnicas Imunoenzimáticas , Imunoglobulina G/análise , Imunoglobulina G/imunologia , Radioisótopos do Iodo/análise , Radioisótopos do Iodo/farmacocinética , Camundongos , Camundongos Nus , Peso Molecular , Transplante de Neoplasias , Polietilenoglicóis/análise , Radioimunodetecção , Distribuição Tecidual , Transplante Heterólogo
11.
J Biol Chem ; 275(18): 13668-76, 2000 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-10788485

RESUMO

The rabbit antibody repertoire, which in the form of polyclonal antibodies has been used in diagnostic applications for decades, would be an attractive source for the generation of therapeutic human antibodies. The humanization of rabbit antibodies, however, has not been reported. Here we use phage display technology to select and humanize antibodies from rabbits that were immunized with human A33 antigen which is a target antigen for the immunotherapy of colon cancer. We first selected rabbit antibodies that bind to a cell surface epitope of human A33 antigen with an affinity in the 1 nm range. For rabbit antibody humanization, we then used a selection strategy that combines grafting of the complementarity determining regions with framework fine tuning. The resulting humanized antibodies were found to retain both high specificity and affinity for human A33 antigen.


Assuntos
Anticorpos/genética , Anticorpos/uso terapêutico , Especificidade de Anticorpos , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Afinidade de Anticorpos , Sequência de Bases , Biblioteca Gênica , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Dados de Sequência Molecular , Coelhos , Especificidade da Espécie
12.
Semin Oncol ; 26(6): 683-90, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10606261

RESUMO

Clinical research in antibody-based cancer therapy has been driven for many years by the prospect of identifying cell-surface antigens with sufficiently restrictive tissue expression patterns to allow the specific targeting of antibody to tumor tissue. Few if any such antibodies capable of targeting rapidly and efficiently to solid tumors have been identified. The main reasons for this are based on the inherent pharmacokinetics and physiology of IgG, the immunoglobulin G molecule. Factors that may limit targeting potential include accessibility of tumor antigen, and antibody affinity, molecular size, and metabolism. Immunoglobulins have evolved to optimally protect an organism from foreign invaders rather than to act as an efficient carrier molecule for therapeutic reagents. Despite these potential limitations, our growing understanding of the biologic and physiologic principles that underlie targeted therapy has led to the development of a generation of novel reagents and the first "positive" clinical trials. Recent strategies for therapeutic use of antibodies in colon cancer have focused on (I) unmodified mouse IgG; (2) immune globulin as carrier for targeted delivery of radioisotopes; toxins, and therapeutic molecules; (3) genetically engineered antibody constructs redesigned for specific uses; (4) humanized, nonimmunogenic IgG structures; and (5) novel antigen targets in tumors. Genetically engineered antibody constructs provide an exciting approach to address and subsequently overcome some of the problems identified for unmodified IgG. These new constructs should increase the dose fraction localized in tumors versus normal tissue and thereby improve the delivery capacity. In contrast, strategies such as immune-mediated cytotoxicity are less dependent on the quantitative difference between the antibody fraction localized in tumor and the nonlocalized fraction. Because antibodies, which direct host cytotoxic mechanisms, become activated in the tumor only when bound to antigen, one would not expect nonspecific toxic effects from nonlocalized antibody. The hypothesis that antibodies alone can destroy tumor tissue solely by directing immune cytotoxic mechanisms is just now being tested in clinical trials evaluating a new generation of humanized antibodies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias do Colo/terapia , Imunotoxinas/uso terapêutico , Radioimunoterapia , Animais , Anticorpos Antineoplásicos , Antígenos de Neoplasias/imunologia , Neoplasias do Colo/imunologia , Humanos , Imunoglobulina G/imunologia , Imunoterapia/métodos
13.
J Nucl Med ; 40(10): 1764-8, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10520720

RESUMO

UNLABELLED: This article compares the effectiveness of radiation delivered by a radiolabeled monoclonal antibody, 131I-labeled A33, that targets colorectal carcinoma, with that of 10 fractions of conventional 320 kVp x-rays. METHODS: Human colorectal cancer xenografts (SW1222) ranging between 0.14 and 0.84 g were grown in nude mice. These were treated either with escalating activities (3.7-18.5 MBq) of 131I-labeled A33 or 10 fractions of 320 kVp x-rays (fraction sizes from 1.5 to 5 Gy). Tumor dosimetry was determined from a similar group of tumor-bearing animals by serial kill, tumor resection and counting of radioactivity in a gamma counter. The relative effectiveness of the two radiation therapy treatment approaches was compared in terms of tumor regrowth delay and probability of tumor cure. RESULTS: The absorbed dose to tumor per MBq administered was estimated as 3.7 Gy (+/-1 Gy; 95% confidence interval). We observed a close to linear increase in tumor regrowth delay with escalating administered activity. Equitumor response of 1311 monoclonal antibody A33 was observed at average radiation doses to the tumor three times greater than when delivered by fractionated external beam radiotherapy. The relationship between the likelihood of tumor cure and administered activity was less predictable than that for regrowth delay. CONCLUSION: The relative effectiveness per unit dose of radiation therapy delivered by 131I-labeled A33 monoclonal antibodies was approximately one third of that produced by fractionated external beam radiotherapy, when measured by tumor regrowth delay.


Assuntos
Neoplasias Colorretais/radioterapia , Radioimunoterapia , Animais , Anticorpos Monoclonais/uso terapêutico , Antígenos de Neoplasias/imunologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Relação Dose-Resposta à Radiação , Feminino , Humanos , Radioisótopos do Iodo/uso terapêutico , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Dosagem Radioterapêutica , Fatores de Tempo , Resultado do Tratamento
14.
Nucl Med Biol ; 26(8): 923-9, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10708306

RESUMO

Monoclonal antibody (MAb) A33 was labeled with the positron emitter 76Br (T(1/2) = 16.2 h). Direct labeling was done using the conventional chloramine-T method. After optimization of the labeling conditions, a maximum yield (mean +/- max error) of 77 +/- 2% was obtained at pH 6.8. In vitro binding of 76Br-A33 to SW1222 colonic cancer cells showed that the immunoreactivity was retained. Also, the MAbs 38S1 and 3S193 and the peptide hEGF were 76Br-labeled, resulting in labeling yields (mean +/- max error) of 75 +/- 3%, 63 +/- 4%, and 73 +/- 0.1%, respectively. We conclude that antibodies and peptides can be labeled conveniently with 76Br for the purpose of whole-body tumour imaging by positron emission tomography.


Assuntos
Anticorpos Monoclonais/química , Cloraminas/química , Compostos Radiofarmacêuticos/síntese química , Compostos de Tosil/química , Anticorpos Monoclonais/imunologia , Radioisótopos de Bromo , Soluções Tampão , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Indicadores e Reagentes , Marcação por Isótopo , Células Tumorais Cultivadas
15.
Clin Cancer Res ; 4(11): 2729-39, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9829736

RESUMO

This Phase I/II radioimmunotherapy study was carried out to determine the maximum tolerated dose (MTD) and therapeutic potential of 131I-G250. Thirty-three patients with measurable metastatic renal cell carcinoma were treated. Groups of at least three patients received escalating amounts of 1311I (30, 45, 60, 75, and 90 mCi/m2) labeled to 10 mg of mouse monoclonal antibody G250, administered as a single i.v. infusion. Fifteen patients were studied at the MTD of activity. No patient had received prior significant radiotherapy; one had received prior G250. Whole-body scintigrams and single-photon emission computed tomography images were obtained in all patients. There was targeting of radioactivity to all known tumor sites that were > or =2 cm. Reversible liver function test abnormalities were observed in the majority of patients (27 of 33 patients). There was no correlation between the amount of 131I administered or hepatic absorbed radiation dose (median, 0.073 Gy/mCi) and the extent or nature of hepatic toxicity. Two of the first six patients at 90 mCi/m2 had grade > or =3 thrombocytopenia; the MTD was determined to be 90 mCi/m2 131I. Hematological toxicity was correlated with whole-body absorbed radiation dose. All patients developed human antimouse antibodies within 4 weeks posttherapy; retreatment was, therefore, not possible. Seventeen of 33 evaluable patients had stable disease. There were no major responses. On the basis of external imaging, 131I-labeled mouse monoclonal antibody G250 showed excellent localization to all tumors that were > or =2 cm. Seventeen of 33 patients had stable disease, with tumor shrinkage observed in two patients. Antibody immunogenicity restricted therapy to a single infusion. Studies with a nonimmunogenic G250 antibody are warranted.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Carcinoma de Células Renais/radioterapia , Imunoconjugados/uso terapêutico , Radioisótopos do Iodo/uso terapêutico , Neoplasias Renais/radioterapia , Adulto , Idoso , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Contagem de Células Sanguíneas/efeitos da radiação , Carcinoma de Células Renais/diagnóstico por imagem , Feminino , Humanos , Imunoconjugados/efeitos adversos , Radioisótopos do Iodo/efeitos adversos , Radioisótopos do Iodo/farmacocinética , Neoplasias Renais/diagnóstico por imagem , Fígado/efeitos dos fármacos , Fígado/efeitos da radiação , Masculino , Pessoa de Meia-Idade , Radioimunoterapia , Radiometria , Cintilografia , Irradiação Corporal Total
16.
Dis Colon Rectum ; 41(9): 1112-5, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9749494

RESUMO

PURPOSE: The purpose of this study was to assess the immunocytochemical status of bone marrow aspirates from patients with clinically isolated hepatic metastases to test the hypothesis that such findings would allow improved patient selection for liver-directed treatment. METHODS: All patients had biopsy-proven or presumed colorectal cancer metastatic to the liver and were scheduled for an operative procedure for hepatic resection or for hepatic artery catheter and chemotherapy pump implant. Immunocytochemical analysis of bone marrow aspirate smears was performed with a panel of monoclonal antibodies directed toward cytokeratins, Lewis Y antigen and A-33 colorectal epitopes. RESULTS: Data from 80 patients indicated that bone marrow reactivity was present in 9.5 percent of those with resectable hepatic metastases and in 34 percent of those not resected (P = 0.03). No single monoclonal antibody or combination produced better discrimination. CONCLUSIONS: Presence or absence of presumed occult colorectal cancer cells in the bone marrow of patients with isolated hepatic metastases is biologically interesting, but not useful in selecting or altering patient management.


Assuntos
Exame de Medula Óssea , Neoplasias da Medula Óssea/secundário , Neoplasias Colorretais/diagnóstico , Neoplasias Hepáticas/secundário , Anticorpos Monoclonais , Biomarcadores Tumorais/análise , Medula Óssea/patologia , Neoplasias da Medula Óssea/diagnóstico , Neoplasias da Medula Óssea/patologia , Neoplasias da Medula Óssea/terapia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Terapia Combinada , Humanos , Técnicas Imunoenzimáticas , Queratinas/análise , Antígenos do Grupo Sanguíneo de Lewis/análise , Fígado/patologia , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Prognóstico
17.
Glycobiology ; 8(9): 927-38, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9675226

RESUMO

We describe a novel immobilization technique to investigate interactions between immobilized gangliosides (GD3, GM1, and GM2) and their respective antibodies, antibody fragments, or binding partners using an optical biosensor. Immobilization was performed by direct injection onto a carboxymethyldextran sensor chip and did not require derivatization of the sensor surface or the ganglioside. The ganglioside appeared to bind to the sensor surface by hydrophobic interaction, leaving the carbohydrate epitope available for antibody or, in the case of GM1, cholera toxin binding. The carboxyl group of the dextran chains on the sensor surface did not appear to be involved in the immobilization as evidenced by equivalent levels of immobilization following conversion of the carboxyl groups into acyl amino esters, but rather the dextran layer provided a hydrophilic coverage of the sensor chip which was essential to prevent nonspecific binding. This technique gave better reactivity and specificity for anti-ganglioside monoclonal antibodies (anti-GD3: KM871, KM641, R24; and anti-GM2: KM966) than immobilization by hydrophobic interaction onto a gold sensor surface or photoactivated cross-linking onto carboxymethydextran. This rapid immobilization procedure has facilitated detailed kinetic analysis of ganglioside/antibody interactions, with the surface remaining viable for a large number of cycles (>125). Kinetic constants were determined from the biosensor data using linear regression, nonlinear least squares and equilibrium analysis. The values of kd, ka, and KAobtained by nonlinear analysis (KAKM871 = 1.05, KM641 = 1.66, R24 = 0.14, and KM966 = 0.65 x 10(7) M-1) were essentially independent of concentration and showed good agreement with data obtained by other analytical methods.


Assuntos
Afinidade de Anticorpos , Especificidade de Anticorpos , Técnicas Biossensoriais , Gangliosídeos/imunologia , Anticorpos Monoclonais , Toxina da Cólera/metabolismo , Gangliosídeo G(M1)/química , Gangliosídeo G(M1)/imunologia , Gangliosídeo G(M2)/química , Gangliosídeo G(M2)/imunologia , Gangliosídeos/química , Análise dos Mínimos Quadrados , Óptica e Fotônica , Propriedades de Superfície
18.
J Chromatogr A ; 798(1-2): 91-101, 1998 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-9542130

RESUMO

Monoclonal antibody (mAb) A33, which recognizes a M(r) approximately 43,000 differentiation antigen (A33) expressed in normal human colonic and small bowel epithelium as well as in 95% of colon cancers, shows specific targeting of colon cancer in humans and is currently being evaluated for clinical use. Here, we describe strategies for the purification and structural analysis of the A33 antigen from the human colorectal carcinoma cell lines LIM1215 and SW1222. Edman degradation of the intact protein and nine peptides, derived by proteolytic digestion of the A33 antigen with Asp-N endoproteinase, thermolysin, trypsin and pepsin followed by micropreparative reversed-phase high-performance liquid chromatography, allowed the unambiguous sequence assignment of 153 amino acid residues; these data reveal one N-glycosylation sequeon in Asp-N endoproteinase peptide D4, and a disulfide linkage between peptides D1 and D4. This amino acid sequence information has facilitated the cloning and subsequent sequencing of a cDNA for the A33 antigen which demonstrates that it is a novel human cell surface molecule of the immunoglobulin superfamily.


Assuntos
Antígenos de Neoplasias/química , Glicoproteínas de Membrana/química , Análise de Sequência/métodos , Sequência de Aminoácidos , Western Blotting , Cromatografia de Afinidade , Cromatografia por Troca Iônica , Neoplasias do Colo , Sequência Conservada , Glicosilação , Humanos , Glicoproteínas de Membrana/isolamento & purificação , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Mapeamento de Peptídeos , Células Tumorais Cultivadas
19.
Int J Oncol ; 12(1): 45-53, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9454885

RESUMO

Monoclonal antibody (mAb) A33 detects a glycoprotein homogeneously expressed by > 95% of human colon cancers and by normal colon cells. The A33 antigen is not secreted or shed and after mAb A33 binds to antigen on the cell membrane, a fraction of membrane-bound mAb A33 is internalized into endosomes. Phase I 131I-mAb A33 biodistribution studies have shown consistent, specific tumor-targeting, and phase I radioimmunotherapy trials with 131I- or 125I-mAb A33 have demonstrated antitumor effects. Here we describe a nude mouse model that was established using a human colon cancer cell line, SW1222, which grows as a relatively hypovascular, invasive heterotransplant when injected i.m. Peak uptake of 131I-labeled or 111In-chelated mAb A33 was observed at 48-96 h, with a mean of 34% (SE +/- 5.0) and 46.7% (SE +/- 1.7) injected dose per gram of tumor tissue, respectively. 111In-mAb A33 was retained in tumor tissue longer than halide radioimmunoconjugates. The specificity of antibody localization was assessed using a control antibody (tumor uptake and pharmacokinetics), a control tumor, corrections for vascular antibody blood-pooling in tumor tissue, and blocking of radiolabeled mAb A33 localization by pretreating mice with excess unlabeled mAb A33. These experiments demonstrate that mAb A33 localization in tumor was specific, and they emphasize the unexpected rapidity with which the antibody localizes. Our conclusions were confirmed by immunohistochemical techniques which allowed direct visualization of localization and distribution of the humanized version of mAb A33 in tumor tissue. Furthermore, antibody doses approximating tumor-saturating doses demonstrated that a homogeneous distribution of antibody in tumor is possible. This model will be valuable for studies focusing on general physiologic aspects of antibody-to-tumor cell localization and critical as a guide to the evaluation of various A33 antibody constructs and combinations with other therapies for the treatment of colon cancer.


Assuntos
Anticorpos Monoclonais/farmacocinética , Anticorpos Antineoplásicos/farmacologia , Antígenos de Neoplasias/imunologia , Neoplasias do Colo/metabolismo , Glicoproteínas de Membrana/imunologia , Neoplasias Experimentais/metabolismo , Animais , Feminino , Humanos , Radioisótopos de Índio , Radioisótopos do Iodo , Camundongos , Camundongos Nus , Radioimunodetecção/métodos , Distribuição Tecidual , Transplante Heterólogo , Células Tumorais Cultivadas
20.
Curr Opin Immunol ; 9(5): 717-22, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9368782

RESUMO

Clinical research in the area of antibody-based tumor-targeted therapy has been driven for many years by the prospect of identifying cell surface antigens with sufficient restrictive tissue expression patterns to allow for the selective and specific accumulation of antibody in tumor tissue. Few, if any, such antibody-antigen systems have been identified which can effectively deliver a large fraction of an administered therapeutic agent to metastatic cancer. Despite this limitation, however, a greater understanding of the biological and physiological principles of tumor-targeted therapy has resulted in successful antibody-based therapy of lymphoma, colon cancer and breast cancer in recent clinical trials.


Assuntos
Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/uso terapêutico , Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...