Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bioconjug Chem ; 30(9): 2393-2403, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31386357

RESUMEN

Following the successful synthesis of a C-functionalized version of the TE1PA ligand, a monopicolinate cyclam, we looked to demonstrate its in vivo properties versus DOTA and NOTA, after conjugation on the 9E7.4 rat antibody, an IgG2a against CD138 murine, which has relevant properties for multiple myeloma targeting. For each ligand, different conjugation approaches had been considered to select the most appropriate for the comparative study. The p-SCN-Bn-TE1PA, NHS-DOTA, and p-SCN-Bn-NOTA were finally chosen for conjugation and radiolabeling tests. For in vivo comparison, we used a model of subcutaneous grafted mice with 5T33 tumor cells. In vitro tests and immuno-PET study highlighted 64Cu-9E7.4-p-SCN-Bn-NOTA as the least attractive. Further competitive biodistribution and hepatic metabolic studies at 2, 24, and 48 h post-injection (100 µg radiolabeled with 10 MBq of 64Cu) were then performed with the 64Cu-9E7.4-p-SCN-Bn-TE1PA and 64Cu-9E7.4-NHS-DOTA. Results show a better in vivo resistance of 64Cu-9E7.4-p-SCN-Bn-TE1PA to transchelation compared to 64Cu-9E7.4-NHS-DOTA, especially at later times. This was confirmed with 64Cu-9E7.4-p-SCN-Bn-NOTA at 48 h PI. 64Cu-9E7.4-p-SCN-Bn-TE1PA also demonstrated an excellent hepatic clearance. 64Cu-9E7.4-p-SCN-Bn-TE1PA displayed an overall superiority compared to 64Cu-9E7.4-NHS-DOTA and 64Cu-9E7.4-p-SCN-Bn-NOTA in terms of in vivo stability, reinforcing the usefulness of the p-SCN-Bn-TE1PA ligand for 64Cu immuno-PET imaging.


Asunto(s)
Anticuerpos Monoclonales/química , Quelantes/química , Radioisótopos de Cobre , Compuestos Heterocíclicos con 1 Anillo/química , Mieloma Múltiple/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Animales , Línea Celular Tumoral , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Ratones , Mieloma Múltiple/patología , Distribución Tisular
2.
Oncotarget ; 9(44): 27502-27513, 2018 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-29938001

RESUMEN

PURPOSE: The aim of this study was to compare the performances pretargeted immunoPET 68Ga-PETimaging (68Ga-pPET) with anti carcino-embryonic antigen (CEA) and anti-histamine-succinyl-glycine (HSG) recombinant humanized bispecific monoclonal antibody (TF2) and 68Ga-labeled HSG peptide (IMP288) to conventional 18FDG-PET in an orthotopic murine model of liver metastases of human colonic cancer. METHODS: Hepatic tumor burden following intra-portal injection of luciferase-transfected LS174T cells in nude mice was confirmed using bioluminescence. One group of animals was injected intravenously with TF2 and with 68Ga-IMP288 24 hours later (n=8). Another group received 18FDG (n=8), and a third had both imaging modalities (n=7). PET acquisitions started 1 hour after injection of the radioconjugate. Biodistributions in tumors and normal tissues were assessed one hour after imaging. RESULTS: Tumor/organ ratios were significantly higher with 68Ga-pPET compared to 18FDG-PET (P<0.05) with both imaging and biodistribution data. 68Ga-pPET sensitivity for tumor detection was 67% vs. 31% with 18FDG PET (P=0.049). For tumors less than 200 mg, the sensitivity was 44% with 68Ga-pPET vs. 0% for 18FDG PET (P=0.031). A strong correlation was demonstrated between tumor uptakes measured on PET images and biodistribution analyses (r2=0.85). CONCLUSION: 68Ga-pPET was more sensitive than 18FDG-PET for the detection of human colonic liver metastases in an orthotopic murine xenograft model. Improved tumor/organ ratios support the use of pretargeting method for imaging and therapy of CEA-expressing tumors.

3.
J Nucl Med ; 54(9): 1597-604, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24003167

RESUMEN

UNLABELLED: New multiple myeloma (MM) treatments--such as high-dose melphalan therapy plus autologous stem cell transplantation or regimens incorporating bortezomide, thalidomide, and lenalidomide--substantially increase the rate of complete response that is associated with longer patient survival. Thus, maintaining the complete response status by improving the minimal residual disease after induction therapy is a key goal for MM management. Here, we address the question of radioimmunotherapy efficacy in MM minimal residual disease treatment in mice with a low tumor burden. α-emitters are particularly well adapted to this approach because the short range of α-particles enables localized irradiation of tumor cells within the bone marrow and a cytotoxic effect on isolated cells due to the high LET (linear energy transfer) of α-particles. The CD138 antigen was used as a target because of its strong expression on myeloma cells in 100% of patients. METHOD: Intravenous injection of 10(6) 5T33 mouse myeloma cells into the Syngeneic mouse strain C57BL/KaLwRij resulted in a rapid invasion of the marrow and limb paralysis, as illustrated by bioluminescence imaging with luciferase-transfected 5T33 cells. Radioimmunotherapy was performed 10 d after 5T33 cell engraftment with an intravenous injection of an antimouse CD138 antibody radiolabeled with (213)Bi at activities of 1.85, 3.7, 7.4, and 11.1 MBq. A blood cell count was performed weekly to monitor hematologic toxicity. The levels of blood Flt3 ligand were also measured to evaluate the radioimmunotherapy-related myelotoxicity. Disease progression was monitored by titrating the monoclonal IgG2b antibody produced by 5T33 cells. RESULTS: The groups treated with 3.7 and 7.4 MBq exhibited a median survival greater than 300 and 227 d, respectively, compared with 45.5 d in the control untreated group. The highest activity (11.1 MBq) showed short-term toxicity whereas the lowest activity (1.85 MBq) gave results similar to those of the controls. With activities of 3.7 and 7.4 MBq, mice exhibited a transient hematologic toxicity whereas only temporary and moderate myelotoxicity was observed with 7.4 MBq. CONCLUSION: This study demonstrates promising therapeutic efficacy of (213)Bi-labeled anti-mCD138 for the treatment of residual disease in the case of MM, with only moderate and transient toxicity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Bismuto/uso terapéutico , Mieloma Múltiple/inmunología , Mieloma Múltiple/radioterapia , Radioinmunoterapia/métodos , Radioisótopos/uso terapéutico , Sindecano-1/inmunología , Animales , Línea Celular Tumoral , Femenino , Ratones , Ratones Endogámicos C57BL , Mieloma Múltiple/diagnóstico , Radiofármacos/uso terapéutico , Tasa de Supervivencia , Resultado del Tratamiento
4.
EJNMMI Res ; 1(1): 20, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22214534

RESUMEN

BACKGROUND: Overexpression of syndecan-1 (CD138) in breast carcinoma correlates with a poor prognosis and an aggressive phenotype. The objective of this study was to evaluate the potential of targeting CD138 by immuno-PET imaging and radioimmunotherapy (RIT) using the antihuman syndecan-1 B-B4 mAb radiolabeled with either 124I or 131I in nude mice engrafted with the triple-negative MDA-MB-468 breast cancer cell line. METHOD: The immunoreactivity of 125I-B-B4 (80%) was determined, and the affinity of 125I-B-B4 and the expression of CD138 on MDA-MB-468 was measured in vitro by Scatchard analysis. CD138 expression on established tumors was confirmed by immunohistochemistry. A biodistribution study was performed in mice with subcutaneous MDA-MB-468 and 125I-B-B4 at 4, 24, 48, 72, and 96 h after injection and compared with an isotype-matched control. Tumor uptake of B-B4 was evaluated in vivo by immuno-PET imaging using the 124I-B-B4 mAb. The maximum tolerated dose (MTD) was determined from mice treated with 131I-B-B4 and the RIT efficacy evaluated. RESULTS: 125I-B-B4 affinity was in the nanomolar range (Kd = 4.39 ± 1.10 nM). CD138 expression on MDA-MB-468 cells was quite low (Bmax = 1.19 × 104 ± 9.27 × 102 epitopes/cell) but all expressed CD138 in vivo as determined by immunohistochemistry. The tumor uptake of 125I-B-B4 peaked at 14% injected dose (ID) per gram at 24 h and was higher than that of the isotype-matched control mAb (5% ID per gram at 24 h). Immuno-PET performed with 124I-B-B4 on tumor-bearing mice confirmed the specificity of B-B4 uptake and its retention within the tumor. The MTD was reached at 22.2 MBq. All mice treated with RIT (n = 8) as a single treatment at the MTD experienced a partial (n = 3) or complete (n = 5) response, with three of them remaining tumor-free 95 days after treatment. CONCLUSION: These results demonstrate that RIT with 131I-B-B4 could be considered for the treatment of metastatic triple-negative breast cancer that cannot benefit from hormone therapy or anti-Her2/neu immunotherapy. Immuno-PET for visualizing CD138-expressing tumors with 124I-B-B4 reinforces the interest of this mAb for diagnosis and quantitative imaging.

5.
J Nucl Med ; 51(4): 624-31, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20351352

RESUMEN

UNLABELLED: A significant antitumor effect was previously observed with radioimmunotherapy using anti-carcinoembryonic antigen (131)I-F6 monoclonal antibody in medullary thyroid cancer-bearing nude mice. Nevertheless, no complete response was observed. As seen with chemotherapy, drugs targeting the tumor microenvironment might improve radioimmunotherapy efficacy. This study evaluated the toxicity and efficacy of combining radioimmunotherapy with thalidomide or a cyclopeptidic vascular endothelial growth inhibitor (CBOP11) in mice grafted with the TT human medullary thyroid cancer cell line. METHODS: Six to 10 nude mice treated with 92.5 MBq of (131)I-F6 in association with 200 mg/kg/d of oral thalidomide during 20 d by force-feeding or 0.45 mg/kg/d of CBOP11 during 25 d using subcutaneous minipumps were compared with control mice receiving either treatment or naked F6 or nonspecific (131)I-734. Combined therapies included (131)I-F6 at day 0 followed by thalidomide between days 20 and 40, thalidomide between days 0 and 20 followed by (131)I-F6 at day 25, (131)I-F6 at day 0 and CBOP11 between days 0 and 25, CBOP11 between days 0 and 25 followed by (131)I-F6 at day 25, and (131)I-F6 at day 0 followed by CBOP11 between days 20 and 45. Animal weight, hematologic toxicity, tumor volume, and serum calcitonin were monitored for the following 3 mo. Improvement of (125)I-F6 tumor biodistribution by antiangiogenic drug was studied after pretreatment by thalidomide. Follow-up of the tumor after combined antiangiogenic and radioimmunotherapy therapies was performed by histology studies. RESULTS: Combined associations, as compared with radioimmunotherapy alone, increased leukopenia but not thrombocytopenia. Tumor volume-quadrupling time (TVQT) was 22.8 +/- 3.3 d in the control group, 29.9 +/- 3.6 d in the group treated with thalidomide, 34.6 +/- 4.4 d in the group treated with CBOP11, and 51.0 +/- 2.8 d after radioimmunotherapy alone. As compared with radioimmunotherapy, TVQT was significantly longer (P < 0.01) after thalidomide followed by radioimmunotherapy (69.83 +/- 3.9), CBOP11 followed by radioimmunotherapy (71.3 +/- 6.1), and CBOP11-radioimmunotherapy in concomitance (64.2 +/- 6.1). Nevertheless, TVQT was not increased after radioimmunotherapy followed by thalidomide (48.8 +/- 4) and radioimmunotherapy followed by CBOP11 (56.8 +/- 4.8). Surprisingly, pretreatment by CBOP11 or thalidomide sensitized larger tumors (>300 mm(3)) to radioimmunotherapy. Change in calcitonin levels confirmed morphologic tumor response. Tumor uptake 24 h after injection of (125)I-F6 was 4.5 +/- 0.6 percentage injected dose per gram (%ID/g) without pretreatment and 8.7 +/- 1.3 %ID/g with pretreatment by thalidomide. An increase of the antitumor effect observed using the antiangiogenic drug combined with radioimmunotherapy was correlated with a decrease of blood vessels shown by von Willebrand immunostaining. CONCLUSION: Pretreatment with antiangiogenic therapies improved radioimmunotherapy efficacy, with acceptable toxicity. Future investigations will be performed to understand how antiangiogenic agents sensitize large tumors to radioimmunotherapy.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antígeno Carcinoembrionario/metabolismo , Regulación Neoplásica de la Expresión Génica , Radioinmunoterapia , Neoplasias de la Tiroides/terapia , Ensayos Antitumor por Modelo de Xenoinjerto , Inhibidores de la Angiogénesis/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Apoptosis/efectos de los fármacos , Antígeno Carcinoembrionario/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Factores de Crecimiento Endotelial/farmacocinética , Factores de Crecimiento Endotelial/farmacología , Factores de Crecimiento Endotelial/uso terapéutico , Humanos , Radioisótopos de Yodo/química , Ratones , Péptidos Cíclicos/farmacocinética , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Talidomida/farmacología , Talidomida/uso terapéutico , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Distribución Tisular , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Cancer ; 116(4 Suppl): 1053-8, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20127950

RESUMEN

BACKGROUND: Significant antitumor effects were previously observed with radioimmunotherapy (RIT) using an anti-carcinoembryonic antigen (CEA) monoclonal antibody (F6) labeled with iodine-131 in medullary thyroid cancer (MTC)-bearing nude mice. Nevertheless, no complete response was achieved. Because angiogenesis is critical for tumor growth, bevacizumab is used to treat solid tumor in clinical practice. The present pilot study evaluated toxicity and efficacy of RIT combined with bevacizumab in mice subcutaneously grafted with TT MTC cells. METHODS: Groups of 4-6 nude mice were treated with 5 microg/g bevacizumab twice weekly during 4 weeks and/or 100 MBq of (131)I-F6. For combined therapy, bevacizumab was given at Day 0 followed by (131)I-F6 at Day 30. The control group received no treatment. Animal weight, hematological toxicity, tumor volume, and serum calcitonin were monitored for 2 or 4 months. RESULTS: Bevacizumab alone induced no cytopenia and no significant weight loss. A weight loss of 12 +/- 1% and 15 +/- 2% was observed in mice treated by RIT alone or bevacizumab + RIT, respectively. RIT alone and combined treatment induced leukopenia and anemia. RIT alone and RIT plus bevacizumab induced tumor responses with minimum relative tumor volume of 0.38 +/- 0.24 and 0.15 +/- 0.07%, respectively, and time to progression of 35 +/- 5 and 56 +/- 11 days, respectively. CONCLUSIONS: Pretreatment with bevacizumab improved RIT efficacy, with similar toxicity as compared as RIT alone.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Carcinoma Medular/terapia , Radioinmunoterapia , Neoplasias de la Tiroides/terapia , Animales , Anticuerpos Monoclonales Humanizados , Bevacizumab , Antígeno Carcinoembrionario/inmunología , Línea Celular Tumoral , Terapia Combinada , Humanos , Ratones , Ratones Desnudos , Radioinmunoterapia/efectos adversos , Factor A de Crecimiento Endotelial Vascular/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Eur J Nucl Med Mol Imaging ; 32(8): 901-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15864584

RESUMEN

PURPOSE: Pretargeted therapy with radiolabelled bivalent haptens and bispecific antibodies has shown promising results, but blood clearance of the activity-carrying haptens under conditions designed for radioimmunotherapy is relatively slow. Thus, the chase of excess circulating bispecific antibody by biotinylation of the bispecific antibody and injection of avidin before hapten administration was tested with a view to increasing tumour-to-blood activity ratios. METHODS: The anti-carcinoembryonic antigen (CEA) x anti-diethylene triamine penta-acetic acid-indium (di-DTPA-indium) bispecific antibody (hMN-14x734) was derivatised with NHS-LC-biotin and injected into LS-174T tumour-bearing nude mice at a dose of 3.5 nmol, followed by avidin and finally by the 125I-labelled di-DTPA-indium hapten (1 nmol). Blood samples were collected, animals sacrificed and tumours and normal tissues counted. RESULTS: Avidin chased up to 72% of the circulating antibody in the liver and the spleen within 30 min. When the labelled hapten was injected 3 h after avidin, tumour to blood ratios measured 3 and 24 h after hapten injection were significantly improved by the chase (3.5-fold), whereas tumour uptake was not significantly reduced. Uptake in normal tissues was unchanged (liver, kidney) or decreased (muscle), with the exception of spleen, in which uptake of both antibody and hapten was increased by the avidin chase. CONCLUSION: The chase strategy reduces hapten concentration in blood and thus should reduce bone marrow exposure. The use of two different recognition systems limits possible interference between the chase and targeting steps.


Asunto(s)
Anticuerpos/sangre , Avidina/administración & dosificación , Antígeno Carcinoembrionario/sangre , Carcinoma/metabolismo , Neoplasias del Colon/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Radioisótopos de Yodo/farmacocinética , Animales , Anticuerpos/inmunología , Anticuerpos/uso terapéutico , Antígeno Carcinoembrionario/uso terapéutico , Carcinoma/radioterapia , Línea Celular Tumoral , Neoplasias del Colon/radioterapia , Combinación de Medicamentos , Femenino , Humanos , Radioisótopos de Yodo/uso terapéutico , Tasa de Depuración Metabólica/efectos de los fármacos , Ratones , Ratones Desnudos , Neoplasias Experimentales , Especificidad de Órganos/efectos de los fármacos , Radioinmunoterapia/métodos , Distribución Tisular/efectos de los fármacos
8.
Mol Cancer Ther ; 1(4): 267-74, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12467222

RESUMEN

A significant antitumor effect associated with moderate toxicity was obtained previously with anticarcinoembryonic antigen x antidiethylene-triaminepentaacetic acid (DTPA)-indium F6-734 bispecific antibody and iodine-131-labeled DTPA-indium bivalent hapten in an animal model of medullary thyroid cancer (MTC). The purpose of this study was to determine whether the cytotoxic agents doxorubicin and paclitaxel, also known as radiosensitizers, improve efficacy of pretargeted radioimmunotherapy (RIT) in experimental MTC. Nude mice bearing TT MTC xenograft were treated with F6-734 and iodine-131-labeled DTPA-indium bivalent hapten injected 48 h apart with or without doxorubicin or paclitaxel. The maximum tolerated dose (MTD) of RIT was 92.5 MBq (as determined previously) and that of doxorubicin and paclitaxel 200 and 1000 micrograms, respectively. A control group received no treatment. Animal weight, hematotoxicity, tumor volume, and serum calcitonin were monitored for 5 months. Tumor growth inhibition induced by drugs alone, RIT alone, or combined therapy was characterized by measuring relative tumor volume 20, 40, and 60 days after treatment to detect additivity or synergism. Mean tumor volume doubling time (MTVDT) was 13 +/- 4 days in the control group, 15 +/- 8 days in the group treated with the MTD of doxorubicin, and 32 +/- 13 days in the group treated with the MTD of paclitaxel. After RIT alone at 92.5 MBq, MTVDT was 86 +/- 22 days. After RIT at 74 MBq (80% of MTD), MTVDT was 56 +/- 10 days. MTVDT was not significantly different from this value after RIT plus doxorubicin, 60 +/- 16 days (65 and 100% of the respective single-agent MTDs). Combination of RIT with paclitaxel (65 and 100% of the respective single-agent MTDs) prolonged the suppression of tumor growth. One complete response was observed, and MTVDT was 114 +/- 44 days. This value was significantly longer than the value obtained with RIT alone at 74 MBq (P < 0.05) or with RIT combined with doxorubicin (P < 0.02). The change in serum calcitonin levels paralleled those in tumor volume. Analysis of dose-response curves at days 20 and 40 showed additivity between RIT and paclitaxel, and analysis at day 60 suggested a synergistic effect. In conclusion, addition of doxorubicin did not improve RIT efficacy, whereas paclitaxel improved RIT efficacy significantly without increasing toxicity.


Asunto(s)
Carcinoma Medular/terapia , Paclitaxel/uso terapéutico , Ácido Pentético/análogos & derivados , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Radioinmunoterapia , Neoplasias de la Tiroides/terapia , Animales , Anticuerpos Biespecíficos , Antígeno Carcinoembrionario/inmunología , Carcinoma Medular/patología , Terapia Combinada , Doxorrubicina/uso terapéutico , Humanos , Inmunoglobulina G/uso terapéutico , Radioisótopos de Yodo/uso terapéutico , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Ácido Pentético/uso terapéutico , Neoplasias de la Tiroides/patología , Trasplante Heterólogo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...