Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
MAbs ; 8(3): 593-603, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26918260

RESUMEN

MPDL3280A is a human monoclonal antibody that targets programmed cell death-1 ligand 1 (PD-L1), and exerts anti-tumor activity mainly by blocking PD-L1 interaction with programmed cell death-1 (PD-1) and B7.1. It is being investigated as a potential therapy for locally advanced or metastatic malignancies. The purpose of the study reported here was to characterize the pharmacokinetics, pharmacodynamics, tissue distribution and tumor penetration of MPDL3280A and/or a chimeric anti-PD-L1 antibody PRO304397 to help further clinical development. The pharmacokinetics of MPDL3280A in monkeys at 0.5, 5 and 20 mg · kg(-1) and the pharmacokinetics / pharmacodynamics of PRO304397 in mice at 1, 3 10 mg · kg(-1) were determined after a single intravenous dose. Tissue distribution and tumor penetration for radiolabeled PRO304397 in tumor-bearing mouse models were determined. The pharmacokinetics of MPDL3280A and PRO304397 were nonlinear in monkeys and mice, respectively. Complete saturation of PD-L1 in blood in mice was achieved at serum concentrations of PRO304397 above ∼ 0.5 µg · mL(-1). Tissue distribution and tumor penetration studies of PRO304397 in tumor-bearing mice indicated that the minimum tumor interstitial to plasma radioactivity ratio was ∼ 0.3; saturation of target-mediated uptake in non-tumor tissues and desirable exposure in tumors were achieved at higher serum concentrations, and the distribution into tumors was dose-and time-dependent. The biodistribution data indicated that the efficacious dose is mostly likely higher than that estimated based on simple pharmacokinetics/pharmacodynamics in blood. These data also allowed for estimation of the target clinical dose for further development of MPDL3280A.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antineoplásicos , Antígeno B7-H1/antagonistas & inhibidores , Puntos de Control del Ciclo Celular , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Experimentales , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/farmacología , Antígeno B7-H1/inmunología , Células CHO , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/inmunología , Cricetinae , Cricetulus , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Macaca fascicularis , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología
2.
J Transl Med ; 13: 191, 2015 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-26070816

RESUMEN

BACKGROUND: Although targeted therapies have improved the clinical outcomes of cancer treatment, tumors resistance to targeted drug are often detected too late and cause mortality. CSE1L is secreted from tumor and its phosphorylation is regulated by ERK1/2. ERK1/2 is located downstream of various growth factor receptors and kinases, the targets of most targeted drugs. Serum phospho-CSE1L may be a marker for monitoring the efficacy of targeted therapy. METHODS: We used mice tumor xenograft model to study the assay of serum phosphorylated CSE1L for early detecting the efficacy of targeted drugs. The phosphorylation status of CSE1L in vemurafenib and sorafenib treated tumor cells were assayed by immunoblotting with antibody against phosphorylated CSE1L. RESULTS: Ras activation increased phospho-CSE1L expression in B16F10 melanoma cells. Vemurafenib and sorafenib treatment did not significantly reduce the total CSE1L levels; however, they inhibited ERK1/2 and CSE1L phosphorylation in A375 melanoma cells and HT-29 colorectal cancer cells. In the melanoma xenograft model, serum phospho-CSE1L level declined 5 days after vemurafenib/sunitinib treatment and 3 days after sorafenib/lapatinib treatment in the HT-29 colon cancer xenograft model. Vemurafenib/sunitinib and sorafenib/lapatinib treatments resulted in tumor regression. CONCLUSIONS: Our results indicated that serum phospho-CSE1L is useful for early detecting the efficacy of targeted therapy in initial treatment and for monitoring emerging secondary drug resistance to facilitate timely therapeutic decision making.


Asunto(s)
Proteína de Susceptibilidad a Apoptosis Celular/sangre , Neoplasias Colorrectales/tratamiento farmacológico , Indoles/uso terapéutico , Melanoma/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Pirroles/uso terapéutico , Quinazolinas/uso terapéutico , Sulfonamidas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Anticuerpos Antineoplásicos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Indoles/farmacología , Lapatinib , Masculino , Melanoma/sangre , Melanoma/patología , Ratones Endogámicos NOD , Ratones SCID , Niacinamida/farmacología , Niacinamida/uso terapéutico , Compuestos de Fenilurea/farmacología , Fosforilación/efectos de los fármacos , Pirroles/farmacología , Quinazolinas/farmacología , Sorafenib , Sulfonamidas/farmacología , Sunitinib , Vemurafenib
3.
J Immunol ; 194(11): 5272-81, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25917091

RESUMEN

In this work, we studied autoantibody repertoires and Ig isotypes in 71 mothers and their 104 healthy newborns (including twins and triplets delivered term or premature). Newborns receive maternal IgG Abs via the placenta before birth, but developing infants must produce their own IgM and IgA Abs. We used an Ag microarray analysis to detect binding to a selection of 295 self-Ags, compared with 27 standard foreign Ags. The magnitude of binding to specific self-Ags was found to be not less than that to the foreign Ags. As expected, each newborn shared with its mother a similar IgG repertoire-manifest as early as the 24th week of gestation. IgM and IgA autoantibody repertoires in cord sera were highly correlated among the newborns and differed from their mothers' repertoires; the latter differed in sera and milk. The autoantibodies bound to self-Ags known to be associated with tumors and to autoimmune diseases. Thus, autoantibody repertoires in healthy humans--the immunological homunculus--arise congenitally, differ in maternal milk and sera, and mark the potential of the immune system to attack tumors, beneficially, or healthy tissues, harmfully; regulation of the tissue site, the dynamics, and the response phenotype of homuncular autoimmunity very likely affects health.


Asunto(s)
Anticuerpos Antineoplásicos/sangre , Autoanticuerpos/sangre , Calostro/inmunología , Sangre Fetal/inmunología , Isotipos de Inmunoglobulinas/sangre , Anticuerpos Antineoplásicos/inmunología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Isotipos de Inmunoglobulinas/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Recién Nacido , Leche Humana/inmunología , Neoplasias/inmunología
4.
Mol Oncol ; 8(2): 378-88, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24389243

RESUMEN

In clinical practice, targeted therapies are usually administered together with chemotherapeutics. However, little is known whether conventional cytotoxic agents enhance the efficacy of targeted compounds, and whether a possible synergy would be dictated by drug-sensitizing genetic alterations. To explore these issues, we leveraged the design of clinical studies in humans to conduct a multi-arm trial in an 'in-cell' format. Using the MET oncogene as a model target and a panel of genetically characterized cell lines as a reference population, we found that two different chemotherapeutic regimens - cisplatin and 5-fluorouracil - exerted widespread cytotoxic activity that was not further enhanced by MET inhibition with a monovalent anti-MET antibody. From a complementary perspective, targeted MET inhibition was successful in a selected complement of cells harboring MET genomic lesions. In this latter setting, addition of chemotherapy did not provide a therapeutic advantage. Mechanistically, chemotherapeutics did not influence the basal activity of MET in cells with normal MET genomic status nor did they contribute to neutralize MET signals in cells with MET amplification. These data suggest that tumors displaying MET aberrations achieve plateau responses by MET monotherapy and do not receive further benefit by addition of cytotoxic treatments.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/farmacología , Citotoxinas/farmacología , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Antineoplásicos/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Cisplatino/farmacología , Citotoxinas/inmunología , Ensayos de Selección de Medicamentos Antitumorales , Fluorouracilo/farmacología , Humanos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Proteínas Proto-Oncogénicas c-met/genética
5.
Int J Cancer ; 135(3): 710-9, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24142484

RESUMEN

Current therapies for glioblastoma are largely palliative, involving surgical resection followed by chemotherapy and radiation therapy, which yield serious side effects and very rarely produce complete recovery. Curcumin, a food component, blocked brain tumor formation but failed to eliminate established brain tumors in vivo, probably because of its poor bioavailability. In the glioblastoma GL261 cells, it suppressed the tumor-promoting proteins NF-κB, P-Akt1, vascular endothelial growth factor, cyclin D1 and BClXL and triggered cell death. Expression of exogenous p50 and p65 subunits of NF-κB conferred partial protection on transfected GL261 cells against curcumin insult, indicating that NF-κB played a key role in protecting glioblastoma cells. To enhance delivery, we coupled curcumin to the glioblastoma-specific CD68 antibody in a releasable form. This resulted in a 120-fold increase in its efficacy to eliminate GL261 cells. A very similar dose response was also obtained with human glioblastoma lines T98G and U87MG. GL261-implanted mice receiving intratumor infusions of the curcumin-CD68 adduct followed by tail-vein injections of solubilized curcumin displayed a fourfold to fivefold reduction in brain tumor load, survived longer, and about 10% of them lived beyond 100 days. Hematoxylin-eosin staining of brain sections revealed a small scar tissue mass in the rescued mice, indicating adduct-mediated elimination of glioblastoma tumor. The tumor cells were strongly CD68+ and some cells in the tumor periphery were strongly positive for microglial Iba1, but weakly positive for CD68. This strategy of antibody targeting of curcumin to tumor comes with the promise of yielding a highly effective therapy for glioblastoma brain tumors.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Curcumina/uso terapéutico , Sinergismo Farmacológico , Glioblastoma/tratamiento farmacológico , Animales , Antígenos CD/química , Antígenos de Diferenciación Mielomonocítica/química , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
6.
Int J Cancer ; 131(4): E569-78, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21989768

RESUMEN

In vitro studies have shown that curcumin, a polyphenol from the culinary component turmeric, has strong anticancer properties. However, there is no consensus on its therapeutic effect in human. Our earlier experiments involving implanted murine melanoma B16F10 cells in the neck or brain of syngeneic C57BL6 mice showed that tail vein injection of curcumin blocks formation of lesions and tumor in these mice. However, such treatment was ineffective in eliminating established tumors that already occupied ≤10% of brain volume. Possible reasons include low solubility and rapid metabolism of curcumin in vivo. To increase its efficacy, we have linked curcumin through a cleavable arm to an antibody (Ab) against the melanoma surface antigen Muc18. The antibody-coupled curcumin was 230-fold more effective in eliminating B16F10 cells in vitro, and in vivo, it rapidly decimated established, B16F10-evoked brain tumors, enabling the rescued mice to live normally far beyond 90 days from implantation of cancer cells. In contrast, mice treated with Muc18 Ab alone died of brain tumor within a month. In B16F10 cells, curcumin-Ab (adduct) treatment caused a dramatic inhibition of NF-kB: a transcription factor that is constitutively activated in cancer cells. Furthermore, overexpression of NF-kB in the B16F10 cells blocked adduct-evoked stimulation of caspase-3/7 activity. Thus, by suppressing NF-kB, the curcumin adduct inhibits other downstream tumor-promoting proteins, thereby eliminating the B16F10 cells. Our study submits a novel yet generally applicable strategy of converting curcumin into a potent anticancer agent and provides a mechanistic framework for its action.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Neoplasias Encefálicas/tratamiento farmacológico , Curcumina/uso terapéutico , Inmunoconjugados/uso terapéutico , Melanoma Experimental/patología , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/secundario , Masculino , Ratones , Ratones Endogámicos C57BL
7.
J Immunol Methods ; 371(1-2): 70-80, 2011 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-21741385

RESUMEN

Carcinoembryonic antigen related cell adhesion molecule (CEACAM) 6 is over-expressed in different types of cancer cells. In addition, it has also been implicated in some infectious diseases. Targeting this molecule by an antibody might have applications in diverse tumor models. Single domain antibody (sdAb) is becoming very useful format in antibody engineering as potential tools for treating acute and chronic disease conditions such as cancer for both diagnostic as well as therapeutic application. Generally, sdAbs with good affinity are isolated from an immune library. Discovery of a new target antigen would require a new immunization with purified antigen which is not always easy. In this study, we have isolated, by phage display, an sdAb against CEACAM6 with an affinity of 5 nM from a llama immunized with cancer cells. The antibody has good biophysical properties, and it binds to the cells expressing the target antigen. Furthermore, it reduces cancer cells proliferation in vitro and shows an excellent tumor targeting in vivo. This sdAb could be useful in diagnosis as well as therapy of CEACAM6 expressing tumors. Finally, we envisage it would be feasible to isolate good sdAbs against other interesting tumor associated antigens from this library. Therefore, this immunization method could be a general strategy for isolating sdAbs against any surface antigen without immunizing the animal with the antigen of interest each time.


Asunto(s)
Anticuerpos Antineoplásicos/aislamiento & purificación , Anticuerpos de Cadena Única/uso terapéutico , Secuencia de Aminoácidos , Animales , Anticuerpos Antineoplásicos/genética , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/uso terapéutico , Afinidad de Anticuerpos , Antígenos CD/administración & dosificación , Antígenos CD/inmunología , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/inmunología , Secuencia de Bases , Camélidos del Nuevo Mundo , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/inmunología , Línea Celular Tumoral , ADN Complementario/genética , Mapeo Epitopo , Proteínas Ligadas a GPI/administración & dosificación , Proteínas Ligadas a GPI/inmunología , Humanos , Inmunización , Datos de Secuencia Molecular , Neoplasias/inmunología , Neoplasias/terapia , Biblioteca de Péptidos , Ingeniería de Proteínas/métodos , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/aislamiento & purificación
8.
Planta Med ; 77(8): 817-24, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21128203

RESUMEN

The 95 % ethanol extract of Astragalus has been demonstrated to have potent activity as an immunological adjuvant when administered with vaccines of various types. We endeavor here to identify the components of this extract that are responsible for this adjuvant activity. Mice were immunized with KLH conjugated to cancer carbohydrate antigens globo H and GD3 and cancer peptide antigen MUC1 combined with different Astragalus fractions or with commercially available Astragalus saponins and flavonoids. The antibody responses against cancer antigens and KLH were quantitated in ELISA assays, and toxicity was calculated by weight loss. Astragalosides II and IV were the most active components, but the toxicity of these two differed dramatically. Astragaloside II was the most toxic Astragalus component with 5-10 % weight loss at a dose of 500 µg while astragaloside IV showed no weight loss at all at this dose, suggesting that astragaloside IV might be utilized as an immunological adjuvant in future studies. Several flavonoids also had significant adjuvant activity. However, when the activities of these known immunologically active components of Astragalus (and of endotoxin) are calculated based on the extent of their presence in the 95 % ethanol extract, they provide only a small proportion of the immunological activity. This raises the possibility that additional uniquely active components of Astragalus may contribute to adjuvant activity, or that the adjuvant activity of Astragalus is greater than the activity of the sum of its parts.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Astragalus propinquus/inmunología , Vacunas contra el Cáncer/farmacología , Flavonoides/farmacología , Extractos Vegetales/farmacología , Saponinas/farmacología , Adyuvantes Inmunológicos/toxicidad , Animales , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/inmunología , Formación de Anticuerpos/efectos de los fármacos , Formación de Anticuerpos/inmunología , Astragalus propinquus/química , Vacunas contra el Cáncer/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Flavonoides/química , Flavonoides/inmunología , Flavonoides/toxicidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Extractos Vegetales/química , Extractos Vegetales/toxicidad , Saponinas/química , Saponinas/inmunología , Saponinas/toxicidad , Triterpenos/química , Triterpenos/inmunología , Triterpenos/farmacología , Triterpenos/toxicidad , Vacunas Conjugadas/inmunología , Vacunas Conjugadas/farmacología
9.
Cancer Immun ; 7: 20, 2007 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-18088084

RESUMEN

Novel therapeutic agents that are safe and effective are needed for the treatment of pancreatic, ovarian, lung adenocarcinomas and mesotheliomas. Mesothelin is a glycosyl-phosphatidyl inositol (GPI)-linked membrane protein of 40 kDa over-expressed in all pancreatic adenocarcinoma and mesothelioma, in >70% of ovarian adenocarcinoma, and in non-small cell lung and colorectal cancers. The biological functions of mesothelin are not known, although it appears to be involved in cell adhesion via its interaction with MUC16. We have recently developed MORAb-009, a mouse-human chimeric IgG1kappa monoclonal antibody with an affinity of 1.5 nM for human mesothelin. Here we provide evidence that MORAb-009 prevents adhesion of mesothelin-bearing tumor cells to MUC16 positive cells and can elicit cell-mediated cytotoxicity on mesothelin-bearing tumor cells. Treatment that included MORAb-009 in combination with chemotherapy led to a marked reduction in tumor growth of mesothelin-expressing tumors in nude mice compared to chemotherapy or MORAb-009 treatment alone. No adverse effects of MORAb-009 were noted during toxicology studies conducted in non-human primates. The preclinical data obtained from our studies warrants pursuing clinical testing of MORAb-009. We have in fact initiated a Phase I clinical study enrolling patients with mesothelin-positive pancreatic, mesothelioma, non-small cell lung and ovarian cancers.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/uso terapéutico , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/inmunología , Neoplasias/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Antineoplásicos/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Endocitosis/efectos de los fármacos , Proteínas Ligadas a GPI , Humanos , Mesotelina , Ratones , Ratones Desnudos , Neoplasias/inmunología
10.
Clin Cancer Res ; 13(18 Pt 2): 5643s-5645s, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875801

RESUMEN

Due to the generally slow and incomplete transit of i.p. infused agents into the circulation, treating disease confined to the peritoneal cavity with chemotherapy, biologics, and/or radionuclides provides a pharmacologic advantage. A higher i.p. concentration can be achieved than could be tolerated by systemic administration. An advantage of i.p. versus i.v. administration for localization of radiolabeled antibodies to small peritoneal surface disease has been shown in animal model and human biopsy studies (1, 2). A recent phase III Gynecologic Oncology Group chemotherapy trial has confirmed a survival advantage for i.p. delivery among women undergoing initial therapy for advanced ovarian cancer (3). Although the therapy was more difficult to tolerate such that 60% of patients randomized to the i.p. arm did not complete the entire regimen, there was a 16-month survival advantage. I.p. radionuclide therapy has been used in treatment of ovarian cancer for more than three decades, but side effects have been problematic in non-tumor-targeted 32P therapy (4). Efforts to improve specificity have used a number of antigens expressed on ovarian cancer cells as targets for selective delivery of radionuclide-conjugates. Mouse models and cell culture have been prominent for preclinical study of agents and strategies in the development of i.p. targeted radionuclide therapy for ovarian cancer. Animal studies, which have directed clinical trials, have shown clear improvement in survival with various modifications including combination chemotherapy, pretargeting, and combination of antibodies over simply delivery of a radiolabeled antibody via i.p. route.


Asunto(s)
Anticuerpos Antineoplásicos/uso terapéutico , Antígenos de Neoplasias/inmunología , Ensayos Clínicos como Asunto , Neoplasias Ováricas/terapia , Neoplasias Peritoneales/terapia , Radioinmunoterapia , Radioisótopos/uso terapéutico , Animales , Anticuerpos Antineoplásicos/inmunología , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Peritoneales/inmunología , Neoplasias Peritoneales/secundario , Radioisótopos/farmacocinética
11.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 24(4): 857-61, 2007 Aug.
Artículo en Chino | MEDLINE | ID: mdl-17899760

RESUMEN

To study pharmacokinetics of injection of iodine-131 labelling MEI-TUO-XI monoclonal antibody (hepatoma monoclonal antibody HAb18 F(ab')2) in vivo. 24 cases of primary hepatocelluar carcinoma (PHC) were equally divided into the low dose group, middle dose group and high dose group. After the relevant injection was administrated into the hepatic artery of each case, intravenous blood and urine samples were separately collected at different time for determination of the radioactive count ratio (min(-1)). The proportion of 131I-HAb18 F(ab')2 in serum of each blood sample was determined, and the radioactive count ratio (min(-1)) of druggery for each blood sample was revised according to the proportion. The pharmacokinetic parameters were calculated using DAS ver 1.0 (Drug And Statistics for Windows) program. The component of urine radiomaterial was determined and the percentages of urine radioactivity in administration dosage were calculated. The catabolism of the injection with time accorded with dynamics two-compartment model. The catabolism product was mainly free-131I and was excreted via kidney; the urine radioactivity was 47.70%-51.16% of administration dosage during 120 h after administration of drug. Therefore, the pharmacokinetics of the injection can satisfy the clinical demands. The drug dose recommended for clinical use was 27.75 MBq of the injection for each kg of human body.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Sistemas de Liberación de Medicamentos , Radioisótopos de Yodo/farmacocinética , Neoplasias Hepáticas/radioterapia , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Antineoplásicos/inmunología , Femenino , Arteria Hepática , Humanos , Fragmentos Fab de Inmunoglobulinas , Inyecciones Intraarteriales , Radioisótopos de Yodo/administración & dosificación , Neoplasias Hepáticas/inmunología , Masculino , Persona de Mediana Edad , Radioinmunoterapia , Adulto Joven
12.
Expert Opin Biol Ther ; 4(7): 1159-65, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15268682

RESUMEN

Recent advances in the molecular and cellular biology of malignancy and tumour immunology have stimulated significant progress in the application of immunotherapies as adjuvant treatments in cancer. Oregovomab (OvaRex, AltaRex) is a murine monoclonal antibody with high affinity to the ovarian cancer associated antigen CA125. Infusion of low-dose antibody results in formation of circulating immune complexes which can trigger a cellular immune response targeting CA125 and the ovarian cancer. Oregovomab has activity following initial chemotherapy and in recurrent disease settings and is in Phase III trials to establish its efficacy to prolong time to relapse in patients with advanced ovarian cancer and favourable outcomes to their front-line treatment. Additional studies of antigen processing and combination chemo-immunotherapy are ongoing. The treatment shows promise as a potential new addition to the standard care of ovarian cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/inmunología , Antígeno Ca-125/inmunología , Inmunoterapia , Neoplasias Ováricas/terapia , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales de Origen Murino , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/inmunología , Afinidad de Anticuerpos , Complejo Antígeno-Anticuerpo/inmunología , Antineoplásicos/uso terapéutico , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Inmunidad Celular , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Inmunoglobulina G/uso terapéutico , Ratones , Ratones SCID , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/inmunología , Recurrencia , Subgrupos de Linfocitos T/inmunología , Resultado del Tratamiento
13.
Int J Oncol ; 25(2): 397-405, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15254737

RESUMEN

Few surrogate markers are available for predicting the survival benefit from chemotherapy in primary breast cancer. We examined tumor growth kinetics by assessing cytokeratin 18 neo-epitope (CK18NE), an apoptosis marker detected by M30 antibody and Ki-67 antigen, a proliferation marker detected by MIB-1 antibody in 72 primary breast cancer patients who underwent pre-operative anthracycline-based chemotherapy. Increase in M30 index and decrease in MIB-1 index after the exposure of 2 to 4 cycles of chemotherapy correlated significantly with pathological tumor response. Univariate survival analysis, conducted in the subgroup of 42 patients who underwent CAF (cyclophosphamide, adriamycin and 5-FU) therapy alone, showed that the patients with the high levels of M30 index (>35 counts/1000 tumor cells) and the low levels of MIB-1 index (<140 counts/1000 tumor cells) after chemotherapy had a remarkably favorable prognosis as compared with patients in other categories. In addition, the alteration in growth kinetics by the treatment showed a significant prognostic value. Multivariate analysis also confirmed that the post-treatment growth kinetics was an independent prognostic indicator. These findings suggest that the alteration in growth kinetics revealed by CK18NE and MIB-1 might be a surrogate marker for predicting the survival benefit from chemotherapy in primary breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Adulto , Anciano , Anticuerpos Antineoplásicos/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Quimioterapia Adyuvante , Ciclofosfamida/uso terapéutico , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Femenino , Fluorouracilo/uso terapéutico , Humanos , Queratinas/análisis , Queratinas/inmunología , Antígeno Ki-67/análisis , Antígeno Ki-67/inmunología , Cinética , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia
14.
Cell Mol Biol (Noisy-le-grand) ; 48(5): 511-6, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12146705

RESUMEN

Animal models are currently used to verify the biodistribution of different radiopharmaceuticals before its clinical application in Nuclear Medicine; however, there may be some limitations. The utilization of labelled anti-tumor monoclonal antibodies (MoAb) in experimental models often requires implant of human antigens (usually a cellular implant), which cannot be achieved in immunocompetent animals. Our purpose was to label an anti-CEA MoAb with technetium-99m (99Tc) and to validate a simplified animal model using a noncellular antigenic implant. MoAb was directly labelled with 99mTc, after reduction with 2-mercaptoethanol. Labeling efficiency was checked by ascending chromatography and immunoreactive fraction was measured in plastic wells sensitized with the antigen. Radiopharmaceutical biodistribution was evaluated by dissection and scintigraphy in 5 mice groups; following the subcutaneous administration of Al(OH)3, CEA adsorbed Al(OH)2 and a control group evaluation. Labeling efficiency was 94+/-3%, which showed to be stable for 24 hr, with immunoreactive fraction above 50%. Invasive biodistribution evaluation showed prolonged blood retention, hepatic and renal uptake. A significant increase in uptake was observed in scintigraphic studies of animals with CEA-adsorbed Al(OH)3 implants compared with the other groups (p<0.05). The non-cellular antigenic implant model simplifies the pre-clinical evaluation of labelled MoAb.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antígeno Carcinoembrionario/inmunología , Tecnecio , Animales , Anticuerpos Antineoplásicos/inmunología , Evaluación Preclínica de Medicamentos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/farmacocinética , Masculino , Ratones , Modelos Animales , Especificidad de Órganos , Cintigrafía , Tecnecio/administración & dosificación , Distribución Tisular
15.
Hum Exp Toxicol ; 21(5): 263-7, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12141397

RESUMEN

GM3 is a ganglioside that has been biochemically identified as dominating the cell surface of several human tumours, but is also found on human normal cells at much lower density. Since GM3 is widely distributed in essentially all types of animal cells, there is a conflict with the concepts of tumour-associated antigen, immunogen, and toxicity. We have designed a GM3-based cancer vaccine for the treatment of human breast and melanoma tumours. Prior to the Phase I clinical trial, we carried out a 12-month dose repeated toxicity study in five male Macaca fascicularis monkeys. Four male monkeys were treated with placebo in a similar way. During the study, no differences were observed between control and treated monkeys related to daily clinical observations (other than local damage) including rectal temperature, blood pressure, respiratory and cardiac rates, weight gain, biochemical and hematological parameters (with the exception of transitory pathological changes), and anti-DNA and anti-nuclear antibodies, although treated monkeys consistently developed both IgM- and IgG-specific anti-GM3 antibodies. Sixty per cent of treated monkeys developed moderate local reactions at the injection site, which disappeared without sequels. We concluded that this GM3 cancer vaccine overcame in monkeys the natural tolerance to GM3 ganglioside evidenced by a strong immune response, while the local reactions elicited-were transitory without apparent important systemic toxicity effects.


Asunto(s)
Vacunas contra el Cáncer/toxicidad , Gangliósido G(M3)/toxicidad , Macaca fascicularis , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antineoplásicos/inmunología , Peso Corporal/efectos de los fármacos , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/prevención & control , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Cromatografía Líquida de Alta Presión , Cromatografía en Capa Delgada , Perros , Evaluación Preclínica de Medicamentos , Gangliósido G(M3)/administración & dosificación , Gangliósido G(M3)/inmunología , Pruebas Hematológicas , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Pruebas de Función Renal , Pruebas de Función Hepática , Masculino , Melanoma/inmunología , Melanoma/prevención & control , Proteolípidos/administración & dosificación , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/prevención & control , Pruebas de Toxicidad
16.
Cancer Res ; 61(6): 2625-31, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11289140

RESUMEN

We have reported previously that s.c. immunization of rats with IL-4 transduced 9L gliosarcoma cells (9L-IL-4) induced a potent antitumor immunity against intracranial, parental 9L tumors. Subcutaneous implantation of 9L-IL-4 influenced the systemic humoral response, which was demonstrated by Th2-type isotype-switching and the induction of cellular immune responses, which played a critical role in the rejection of tumors. Serological analyses of recombinant cDNA expression libraries (SEREX), has recently emerged as a powerful method for serological identification of tumor-associated antigens (TAAs) and/or tumor rejection antigens (TRAs). Because IL-4 is known to activate B cells and to promote humoral responses, and inasmuch as induction of humoral responses by central nervous system tumors has been reported to be minimal, we investigated whether the induction of a potent humoral immune response against 9L TAAs or TRAs in rats immunized s.c. with 9L-IL4 could be demonstrated. Screening of 5 x 10(5) independent clones of 9L-expression cDNA library for the presence of reactive antibodies in the serum from a 91-IL-4 immunized rat led to the identification of three different TAAs. One 9L TAA (clone 29) was demonstrated to be calcyclin, a member of the S-100 family of calcium-binding proteins. The second 9L TAA (clone 37) was demonstrated to be the rat homologue of the J6B7 mouse immunomodulatory molecule. The third TAA (clones 158 and 171) was determined to be the rat homologue of the mouse Id-associated protein 1 (MIDA1), a DNA-binding, protein-associated protein. Northern blotting demonstrated that message for calcyclin was overexpressed in 9L cells. Message encoding MIDA1 was highly expressed in parental 9L cells and thymus and, to a lesser degree, in testis, suggesting that MIDA1 was comparable with the cancer/testis category of TAAs. Sera obtained from animals bearing 9L-IL-4 were found to have a higher a frequency and titer of antibodies to these antigens when compared with sera obtained from rats bearing sham-transduced 9L (9L-neo) cells. To determine whether immunization with these TAAs induced antitumor immunity, animals were immunized by intradermal injection with expression plasmids encoding calcyclin or MIDA1. Subsequent challenge of rats with parental 9L resulted in significant suppression of tumor growth in animals immunized with MIDA1, but not with calcyclin. These results indicate that MIDA1 is an effective 9L TRA and will be useful for the investigation of specific antitumor immunity in this glioma model. Furthermore, these results suggest that this approach, termed "cytokine-assisted SEREX (CAS)," may serve as an effective strategy for identification of TRAs for in animal-glioma models of cytokine gene therapy, and potentially in humans undergoing cytokine gene therapy protocols as well.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Proteínas de Ciclo Celular , Gliosarcoma/inmunología , Pruebas Serológicas/métodos , Vacunas de ADN/inmunología , Animales , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/inmunología , Antígenos de Neoplasias/sangre , Antígenos de Neoplasias/aislamiento & purificación , Secuencia de Bases , Vacunas contra el Cáncer/genética , División Celular/inmunología , ADN Complementario/administración & dosificación , ADN Complementario/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Gliosarcoma/patología , Isotipos de Inmunoglobulinas/inmunología , Región de Cambio de la Inmunoglobulina/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Masculino , Ratones , Datos de Secuencia Molecular , Ratas , Ratas Endogámicas F344 , Proteína A6 de Unión a Calcio de la Familia S100 , Proteínas S100/genética , Proteínas S100/inmunología , Sensibilidad y Especificidad , Células Th2/inmunología , Células Tumorales Cultivadas , Vacunas de ADN/genética
17.
Annu Rev Immunol ; 19: 1-21, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11244028

RESUMEN

One of the hallmarks of the immune system is specificity, a concept based on innumerable observations that antibodies react with the substance that elicited their production and only a few other structurally similar substances. The study of T cells has begun to suggest, however, that in responses mediated by their antibody-like receptors (T cell receptor or TCR) an individual T cell, expressing a singular TCR, can discriminate as exquisitely among antigens as the most specific antibodies but also exhibit "degeneracy": i.e., it can react with many disparate antigens (peptide-MHC complexes). An explanation for this duality (specificity and degeneracy) can be found in (i) the powerful amplifying signal transduction cascades that allow a T cell to respond to the stable engagement of very few TCR molecules, initially perhaps only one or two out of around 100,000 per cell, by their natural ligands (peptide-MHC complexes or epitopes on antigen-presenting cells--or APC) and (ii) the inverse relationship between TCR affinity for epitopes and epitope density (the number of copies of an epitope per APC). Older observations on the excess of total globulin production over specific antibody production in response to conventional immunization procedures suggest that B cells also exhibit degeneracy, as well as specificity. These views are developed against a backdrop describing how the author became interested in the immune system and has pursued that interest. "...a concept of science drawn from ...is [textbooks]...is no more likely to fit the enterprise that produced them than an image of a national culture drawn from a tourist brochure." Thomas Kuhn, Structure Of Scientific Revolutions


Asunto(s)
Alergia e Inmunología/historia , Reacciones Antígeno-Anticuerpo/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos Antineoplásicos/inmunología , Especificidad de Anticuerpos , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Sitios de Unión de Anticuerpos , Boston , Haptenos/inmunología , Historia del Siglo XX , Humanos , Hipersensibilidad Tardía/inmunología , Ratones , Missouri , Modelos Inmunológicos , Proteínas de Mieloma/inmunología , Ciudad de Nueva York , Subgrupos de Linfocitos T/inmunología
18.
Cytotherapy ; 3(3): 197-201, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-12171726

RESUMEN

BACKGROUND: We have investigated a novel nonmyeloablative conditioning regimen in 44 patients with hematological malignancies. The median patient age was 41 years. Many of the patients had high-risk features, including 19 patients with a previous failed transplant. METHODS: Recipient conditioning consisted of CAMPATH-1H 20 mg/day on Days -8 to -4, fludarabine 30 mg/m(2) on Days -7 to -3 and melphalan 140 mg/m(2) on Day -2. Thirty-six recipients received unmanipulated G-CSF mobilized PBSC from HLA identical siblings and eight received unmanipulated BM from MUD. GvHD prophylaxis was with CYA alone for 38 patients and CYA plus MTX for six sibling recipients. RESULTS: Forty-two of the 43 evaluable patients had sustained engraftment. Results of chimerism analysis using microsatellite PCR indicate that 18 of 31 patients studied were full donor chimeras, while the other patients were mixed chimeras in one or more lineages. At a median follow-up of 9 months (range, 3-29 months) 33 patients remain alive in CR, or with no evidence of disease progression. Seven patients relapsed or progressed post-transplant and four of them subsequently died. Four patients died from regimen-related complications. There were no cases of Grades III-IV acute GvHD. Only two patients developed Grade II acute GvHD and only one had chronic GvHD. The estimated probability of non-relapse mortality at 1 year was 11%.Results: Although longer follow-up is needed to establish the long-term remission rates, this study demonstrates that this nonmyeloablative preparative regimen is associated with durable engraftment, minimal toxicity and low incidence of GvHD.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/prevención & control , Neoplasias Hematológicas/terapia , Terapia de Inmunosupresión/métodos , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/métodos , Acondicionamiento Pretrasplante/métodos , Vidarabina/análogos & derivados , Adolescente , Adulto , Alemtuzumab , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Anticuerpos Antineoplásicos/inmunología , Antineoplásicos Alquilantes/uso terapéutico , Quimioterapia Combinada , Femenino , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Enfermedad Injerto contra Huésped/inmunología , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/fisiopatología , Humanos , Terapia de Inmunosupresión/tendencias , Inmunosupresores/uso terapéutico , Masculino , Melfalán/uso terapéutico , Persona de Mediana Edad , Recurrencia , Tasa de Supervivencia , Quimera por Trasplante/inmunología , Acondicionamiento Pretrasplante/tendencias , Trasplante Homólogo/efectos adversos , Trasplante Homólogo/métodos , Resultado del Tratamiento , Vidarabina/uso terapéutico
19.
J Exp Med ; 191(11): 1945-56, 2000 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-10839809

RESUMEN

One major objective of tumor immunologists is to prevent cancer development in individuals at high risk. (TG.AC x C57BL/6)F1 mice serve as a model for testing the feasibility of this objective. The mice carry in the germline a mutant ras oncogene that has an arginine at codon 12 instead of glycine present in the wild-type, and after physical (wounding) or chemical promotion, these mice have a high probability for developing papillomas that progress to cancer. Furthermore, F1 mice immunized with Arg(12) mutant ras peptide in complete Freund's adjuvant (CFA) develop T cells within 10 d that proliferate in vitro on stimulation with the Arg(12) mutant ras peptide. Within 14 d, these mice have delayed-type hypersensitivity to the peptide. Immunization with CFA alone or with a different Arg(12) mutant ras peptide in CFA induced neither response. To determine the effect of immunization on development of tumors, mice immunized 3 wk earlier were painted on the back with phorbol 12-myristate 13-acetate every 3 d for 8 wk. The time of appearance and the number of papillomas were about the same in immunized and control mice, but the tumors grew faster and became much larger in the mice immunized with the Arg(12) mutant ras peptide. Thus, the immunization failed to protect against growth of papillomas. The peptide-induced CD4(+) T cells preferentially recognized the peptide but not the native mutant ras protein. On the other hand, mice immunized with Arg(12) mutant ras peptide and bearing papillomas had serum antibodies that did bind native mutant ras protein. Together, these studies indicate that active immunization of cancer-prone individuals may result in immune responses that fail to eradicate mutant oncogene-expressing tumor cells, but rather induce a remarkable enhancement of tumor growth.


Asunto(s)
Arginina/inmunología , Glicina/inmunología , Proteína Oncogénica p21(ras)/inmunología , Papiloma/inmunología , Mutación Puntual , Animales , Anticuerpos Antineoplásicos/inmunología , Arginina/genética , Vacunas contra el Cáncer/inmunología , Femenino , Glicina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Oncogénica p21(ras)/genética , Papiloma/fisiopatología , Péptidos/inmunología , Vacunación
20.
Clin Cancer Res ; 5(5): 953-61, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10353726

RESUMEN

The tumor-associated glycoprotein 72 (TAG-72) antigen is present on a high percentage of tumor types including ovarian carcinomas. Antibody B72.3 is a murine monoclonal recognizing the surface domain of the TAG-72 antigen and has been widely used in human clinical trials. After our initial encouraging studies (M. G. Rosenblum et al., J. Natl. Cancer Inst., 83: 1629-1636, 1991) of tissue disposition, metabolism, and pharmacokinetics in 9 patients with ovarian cancer, we designed an escalating dose, multi-arm Phase I study of 90Y-labeled B72.3 i.p. administration. In the first arm of the study, patients (3 pts/dose level) received an i.p. infusion of either 2 or 10 mg of B72.3 labeled with either 1, 10, 15, or 25 mCi of 90Y. Pharmacokinetic studies demonstrated that concentrations of 90Y-labeled B72.3 persist in peritoneal fluid with half-lives >24 h after i.p. administration. In addition, 90Y-labeled B72.3 was absorbed rapidly into the plasma with peak levels achieved within 48 h, and levels declined slowly thereafter. Cumulative urinary excretion of the 90Y label was 10-20% of the administered dose which suggests significant whole-body retention of the radiolabel. Biopsy specimens of bone and marrow obtained at 72 h after administration demonstrated significant content of the label in bone (0.015% of the dose/g) with relatively little in marrow (0.005% of the dose/g). The maximal tolerated dose was determined to be 10 mCi because of hematological toxicity and platelet suppression. This typically occurred on the 29th day after administration and was thought to be a consequence of the irradiation of the marrow from the bony deposition of the radiolabel. In an effort to suppress the bone uptake of 90Y, patients were treated with a continuous i.v. infusion of EDTA (25 mg/kg/12 h x 6) infused immediately before i.p. administration of the radiolabeled antibody. Patients (3 pts/dose level) were treated with doses of 10, 15, 20, 25, 30, 35, 40, or 45 mCi of 90Y-labeled B72.3 for a total of 38 patients. EDTA administration resulted in significant myeloprotection, which allowed escalation to the maximal tolerated dose of 40 mCi. Dose-limiting toxicity was thrombocytopenia and neutropenia. Studies of plasma and peritoneal fluid pharmacokinetics demonstrate no changes compared with patients without EDTA pretreatment. Cumulative urinary excretion of the radiolabel was not increased in patients pretreated with EDTA compared with the untreated group. However, analysis of biopsy specimens of bone and marrow demonstrated that bone and marrow content of the 90Y label was 15-fold lower (<0.001% injected dose/g) than a companion group without EDTA. Four responses were noted in patients who received 15-30 mCi of 90Y-labeled B72.3 with response durations of 1-12 months. These results demonstrate the myeloprotective ability of EDTA, which allows safe i.p. administration of higher doses of 90Y-labeled B72.3 and, therefore, clearly warrant an expanded Phase II trial in patients with minimal residual disease after standard chemotherapy or for the palliation of refractory ascites.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/uso terapéutico , Antígenos de Neoplasias/inmunología , Enfermedades de la Médula Ósea/prevención & control , Carcinoma/radioterapia , Quelantes/uso terapéutico , Ácido Edético/uso terapéutico , Glicoproteínas/inmunología , Neoplasias Ováricas/radioterapia , Traumatismos por Radiación/prevención & control , Radioinmunoterapia , Radioisótopos/uso terapéutico , Iterbio/uso terapéutico , Adulto , Anciano , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Antineoplásicos/administración & dosificación , Anticuerpos Antineoplásicos/inmunología , Ascitis/radioterapia , Líquido Ascítico/química , Médula Ósea/química , Médula Ósea/efectos de la radiación , Enfermedades de la Médula Ósea/inducido químicamente , Huesos/química , Carcinoma/patología , Carcinoma/secundario , Carcinoma/terapia , Terapia por Quelación , Relación Dosis-Respuesta Inmunológica , Relación Dosis-Respuesta en la Radiación , Ácido Edético/administración & dosificación , Ácido Edético/farmacología , Neoplasias de las Trompas Uterinas/patología , Neoplasias de las Trompas Uterinas/radioterapia , Neoplasias de las Trompas Uterinas/terapia , Femenino , Semivida , Humanos , Inyecciones Intraperitoneales , Ratones , Persona de Mediana Edad , Neoplasia Residual , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Neoplasias Peritoneales/radioterapia , Neoplasias Peritoneales/secundario , Neoplasias Peritoneales/terapia , Traumatismos por Radiación/inducido químicamente , Radioinmunoterapia/efectos adversos , Radioisótopos/administración & dosificación , Radioisótopos/efectos adversos , Radioisótopos/farmacocinética , Dosificación Radioterapéutica , Distribución Tisular , Resultado del Tratamiento , Iterbio/administración & dosificación , Iterbio/efectos adversos , Iterbio/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA