Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Target Oncol ; 13(2): 189-203, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29423595

RESUMO

Alpha-emitters are radionuclides that decay through the emission of high linear energy transfer α-particles and possess favorable pharmacologic profiles for cancer treatment. When coupled with monoclonal antibodies, peptides, small molecules, or nanoparticles, the excellent cytotoxic capability of α-particle emissions has generated a strong interest in exploring targeted α-therapy in the pre-clinical setting and more recently in clinical trials in oncology. Multiple obstacles have been overcome by researchers and clinicians to accelerate the development of targeted α-therapies, especially with the recent improvement in isotope production and purification, but also with the development of innovative strategies for optimized targeting. Numerous studies have demonstrated the in vitro and in vivo efficacy of the targeted α-therapy. Radium-223 (223Ra) dichloride (Xofigo®) is the first α-emitter to have received FDA approval for the treatment of prostate cancer with metastatic bone lesions. There is a significant increase in the number of clinical trials in oncology using several radionuclides such as Actinium-225 (225Ac), Bismuth-213 (213Bi), Lead-212 (212Pb), Astatine (211At) or Radium-223 (223Ra) assessing their safety and preliminary activity. This review will cover their therapeutic application as well as summarize the investigations that provide the foundation for further clinical development.


Assuntos
Partículas alfa/uso terapêutico , Neoplasias/terapia , Humanos
2.
EJNMMI Res ; 6(1): 83, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27873240

RESUMO

BACKGROUND: Targeted alpha therapy (TAT) offers advantages over current ß-emitting conjugates for peptide receptor radionuclide therapy (PRRT) of neuroendocrine tumors. PRRT with 177Lu-DOTATATE or 90Y-DOTATOC has shown dose-limiting nephrotoxicity due to radiopeptide retention in the proximal tubules. Pharmacological protection can reduce renal uptake of radiopeptides, e.g., positively charged amino acids, to saturate in the proximal tubules, thereby enabling higher radioactivity to be safely administered. The aim of this preclinical study was to evaluate the therapeutic effect of 213Bi-DOTATATE with and without renal protection using L-lysine in mice. Tumor uptake and kinetics as a function of injected mass of peptide (range 0.03-3 nmol) were investigated using 111In-DOTATATE. These results allowed estimation of the mean radiation absorbed tumor dose for 213Bi-DOTATATE. Pharmacokinetics and dosimetry of 213Bi-DOTATATE was determined in mice, in combination with renal protection. A dose escalation study with 213Bi-DOTATATE was performed to determine the maximum tolerated dose (MTD) with and without pre-administration of L-lysine as for renal protection. Neutrophil gelatinase-associated lipocalin (NGAL) served as renal biomarker to determine kidney injury. RESULTS: The maximum mean radiation absorbed tumor dose occurred at 0.03 nmol and the minimum at 3 nmol. Similar mean radiation absorbed tumor doses were determined for 0.1 and 0.3 nmol with a mean radiation absorbed dose of approximately 0.5 Gy/MBq 213Bi-DOTATATE. The optimal mass of injected peptide was found to be 0.3 nmol. Tumor uptake was similar for 111In-DOTATATE and 213Bi-DOTATATE at 0.3 nmol peptide. Lysine reduced the renal uptake of 213Bi-DOTATATE by 50% with no effect on the tumor uptake. The MTD was <13.0 ± 1.6 MBq in absence of L-lysine and 21.7 ± 1.9 MBq with L-lysine renal protection, both imparting an LD50 mean renal radiation absorbed dose of 20 Gy. A correlation was found between the amount of injected radioactivity and NGAL levels. CONCLUSIONS: The therapeutic potential of 213Bi-DOTATATE was illustrated by significantly decreased tumor burden and improved overall survival. Renal protection with L-lysine immediately prior to TAT with 213Bi-DOTATATE prolonged survival providing substantial evidence for pharmacological nephron blockade to mitigate nephrotoxicity.

3.
Biomedicines ; 4(1)2015 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-28536369

RESUMO

BACKGROUND: Radioimmunotherapy utilize a targeting antibody coupled to a therapeutic isotope to target and treat a tumor or disease. In this study we examine the synthesis and cell binding of a polymer scaffold containing a radiotherapeutic isotope and a targeting antibody. METHODS: The multistep synthesis of a fluorescent or 149Promethium-labeled Trastuzumab-polyethyleneimine (PEI), Trastuzumab, or PEI is described. In vitro uptake, internalization and/or the binding affinity to the Her2/neu expressing human breast adenocarcinoma SKBr3 cells was investigated with the labeled compounds. RESULTS: Fluorescent-labeled Trastuzumab-PEI was internalized more into cells at 2 and 18 h than fluorescent-labeled Trastuzumab or PEI. The fluorescent-labeled Trastuzumab was concentrated on the cell surface at 2 and 18 h and the labeled PEI had minimal uptake. DOTA-PEI was prepared and contained an average of 16 chelates per PEI; the compound was radio-labeled with 149Promethium and conjugated to Trastuzumab. The purified 149Pm-DOTA-PEI-Trastuzumab had a radiochemical purity of 96.7% and a specific activity of 0.118 TBq/g. The compound demonstrated a dissociation constant for the Her2/neu receptor of 20.30 ± 6.91 nM. CONCLUSION: The results indicate the DOTA-PEI-Trastuzumab compound has potential as a targeted therapeutic carrier, and future in vivo studies should be performed.

4.
ACS Chem Biol ; 5(7): 681-90, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20486699

RESUMO

Recent clinical studies implicate the role of G protein-coupled estrogen receptor, GPR30, in aggressive forms of breast, ovarian, and endometrial cancers. However, the functional role of GPR30 at cellular and molecular levels remains less clear and controversial, particularly its subcellular location. The primary objective of this study was to develop radiolabeled neutral and charged GPR30-targeted nonsteroidal analogues to understand the influence of ligand charge on cell binding, cellular permeability, and in vivo tumor imaging. Therefore, we developed a series of GPR30-targeted (111/113)In(III)-labeled analogues using macrocyclic and acyclic polyamino-polycarboxylate chelate designs that would render either a net negative or neutral charge. In vitro biological evaluations were performed to determine the role of negatively charged analogues on receptor binding and activation using calcium mobilization and phosphoinositide 3-kinase assays. In vivo evaluations were performed on GPR30-expressing human endometrial Hec50 tumor-bearing mice to characterize the biodistribution and potential application of GPR30-targeted imaging agents for translational research. In vitro functional assays revealed an effect of charge, such that only the neutral analogue activated GPR30-mediated rapid signaling pathways. These observations are consistent with expectations for initial rates of membrane permeability and suggest an intracellular rather than the cell surface location of functional receptor. In vivo studies revealed receptor-mediated uptake of the radiotracer in target organs and tumors; however, further structural modifications will be required for the development of future generations of GPR30-targeted imaging agents with enhanced metabolic properties and decreased nonspecific localization to the intestines.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Neoplasias Uterinas/metabolismo , Animais , Ligação Competitiva , Neoplasias da Mama/patologia , Condutividade Elétrica , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Ativação Enzimática , Estrogênios/metabolismo , Feminino , Humanos , Radioisótopos de Índio , Camundongos , Camundongos Knockout , Neoplasias Ovarianas/patologia , Permeabilidade , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Distribuição Tecidual , Neoplasias Uterinas/patologia
8.
Nucl Med Biol ; 35(6): 673-8, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18678352

RESUMO

INTRODUCTION: Clinical studies of patients treated with somatostatin-receptor (sstr)-targeted [DOTA(0)-Tyr(3)]-octreotide (DOTATOC) labeled with (177)Lu and (90)Y have shown overall response rates in the range of 9-33%. This study evaluates the potential for combination therapy with gemcitabine in an effort to improve clinical outcomes. METHODS: Human pancreatic adenocarcinoma Capan-2, rat pancreatic cancer AR42J and human small cell lung cancer NCI-H69 cells were each treated with 1 microg/ml gemcitabine for 4 days followed by replacement of the medium alone for four additional days. Cell cycle and direct receptor-uptake studies were performed with (177)Lu-DOTATOC after the total 8-day treatment as described. Cell viability and apoptosis experiments were performed to study the effects of gemcitabine pretreatment and (177)Lu-DOTATOC radionuclide therapy. Parallel control studies were performed with receptor-non-targeted (177)Lu-DOTA and DOTATOC. RESULTS: Cells treated with gemcitabine for 4 days showed a down-regulation of sstr expression as determined by (177)Lu-DOTATOC uptake. However, after 4 days of additional growth in absence of gemcitabine, the uptake of (177)Lu-DOTATOC was 1.5-3 times greater than that of the untreated control cells. In gemcitabine-pretreated Capan-2 cells, 84% of the cell population was in the G(2)M phase of the cell cycle. Due to sstr up-regulation and cell cycle modulations, synergistic effects of gemcitabine pretreatment were observed in cell viability and apoptosis assays. (177)Lu-DOTATOC resulted in two to three times greater apoptosis in gemcitabine-pretreated Capan-2 cells compared to the untreated cells. CONCLUSION: Gemcitabine pretreatment up-regulates sstr expression and acts as a radiosensitizer through cell cycle modulation. The rational combination of gemcitabine and sstr-targeted radiopharmaceuticals represents a promising chemoradiation therapeutic tool with great potential to improve clinical outcomes and, thus, merits further study.


Assuntos
Ciclo Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Desoxicitidina/análogos & derivados , Neoplasias/patologia , Neoplasias/fisiopatologia , Octreotida/análogos & derivados , Compostos Radiofarmacêuticos/administração & dosagem , Receptores de Somatostatina/efeitos da radiação , Antimetabólitos Antineoplásicos/administração & dosagem , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Humanos , Octreotida/administração & dosagem , Octreotida/uso terapêutico , Radiossensibilizantes/administração & dosagem , Compostos Radiofarmacêuticos/uso terapêutico , Receptores de Somatostatina/antagonistas & inibidores , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/efeitos da radiação , Gencitabina
10.
Nucl Med Biol ; 34(2): 185-93, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17307126

RESUMO

INTRODUCTION: Advance clinical cancer therapy studies of patients treated with somatostatin receptor (sstr)-targeted [DOTA(0)-Tyr(3)]octreotide (DOTATOC) labeled with low-linear-energy-transfer (LET) beta(-)-emitters have shown overall response rates in the range of 15-33%. In order to improve outcomes, we sought to compare the therapeutic effectiveness of sstr-targeted high-LET alpha-emitting (213)Bi to that of low-LET beta(-)-emitting (177)Lu by determining relative biological effectiveness (RBE) using the external gamma-beam of (137)Cs as reference radiation. METHODS: Sstr-expressing human pancreatic adenocarcinoma Capan-2 cells and A549 control cells were used for this study. The effects of different radiation doses of (213)Bi and (177)Lu labeled to 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid and sstr-targeted DOTATOC were investigated with a clonogenic cell survival assay. Apoptosis was measured using the Cell Death Detection ELISA(PLUS) 10x kit. RESULTS: Using equimolar DOTATOC treatment with concurrent irradiation with a (137)Cs source as reference radiation, the calculated RBE of [(213)Bi]DOTATOC was 3.4, as compared to 1.0 for [(177)Lu]DOTATOC. As measured in terms of absorbance units, [(213)Bi]DOTATOC caused a 2.3-fold-greater release of apoptosis-specific mononucleosomes and oligonucleosomes than [(177)Lu]DOTATOC at the final treatment time of 96 h (P<.001) in sstr-expressing Capan-2 cells. CONCLUSIONS: In conclusion, at the same absorbed dose, [(213)Bi]DOTATOC is therapeutically more effective in decreasing survival than is [(177)Lu]DOTATOC in human pancreatic adenocarcinoma cells due to its comparatively higher RBE.


Assuntos
Adenocarcinoma/radioterapia , Bismuto/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Lutécio/administração & dosagem , Neoplasias Pancreáticas/radioterapia , Radioisótopos/administração & dosagem , Receptores de Somatostatina/antagonistas & inibidores , Adenocarcinoma/patologia , Partículas alfa/uso terapêutico , Partículas beta/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Humanos , Neoplasias Pancreáticas/patologia , Compostos Radiofarmacêuticos/administração & dosagem , Dosagem Radioterapêutica , Resultado do Tratamento
11.
Nucl Med Biol ; 32(7): 741-7, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16243650

RESUMO

OBJECTIVE: Pretargeting is the concept that combines optimal delivery of the antibody and rapid capture and elimination of the radioactivity. In this study, we evaluated the potential of antibody pretargeting to enable the tumor-targeting (212)Pb for in vivo generation of (212)Bi for alpha particle radiotherapy. METHODS: The (212)Pb/(212)Bi chelate of DOTA-biotin, as well as their gamma-emitting analogues, (203)Pb and (205)Bi, was prepared and characterized. The radiolabeled compounds were injected in animals for evaluation of tumor targeting and normal tissue uptake and retention. In the pretargeting protocol, injection of 400 microg of NR-LU-10 antibody-streptavidin conjugate was given at t = 0 h, then 100 microg of N-acetyl-galatosamine-biotin clearing agent was injected at t = 20-24 h; finally, 1 microg of (212)Pb/(212)Bi-DOTA-biotin was injected 6 h later. RESULTS: Both (203)Pb and (205)Bi-DOTA-biotin were stable for at least 4 days in the different challenging solutions including PBS, 10 mM DTPA and serum. Contrary to its gamma-emitting analogues, radiolabeled (212)Pb-DOTA-biotin was not stable. There was greater than 30% of free (212)Bi released 4 h after (212)Pb-labeled DOTA-biotin. The results of pretargeting protocol of (203)Pb and (205)Bi-DOTA-biotin showed that the tumor target reached 20% injected dose (ID)/g at 4 h postinjection and remained high for 5 days. The %ID/g in the whole blood and other nontarget organs was low after administration of labeled (203)Pb and (205)Bi-DOTA-biotin similar to the biodistribution of labeled DOTA-biotin alone. In the animals administered (212)Pb-DOTA-biotin, radioactivity in nontarget organs was low except the kidneys. The %ID/g in the kidney for (212)Bi was 14.5 at 2 h, higher than (212)Pb, but dropped to about 6% ID/g by 4 h. However, tumor uptake for (212)Pb and (212)Bi was >25% ID/g at 1 h postinjection and remained so through 24 h. CONCLUSIONS: Antibody pretargeting system with Mab-streptavidin, clearing agent and DOTA-biotin provides the potential of (212)Bi for solid tumor radiotherapy despite the release of (212)Bi after (212)Pb decay. Dosimetry calculations resulted in tumor dose at 93 rad/muCi and ratios of tumor to marrow and kidney at 386:1 and 12:1, respectively.


Assuntos
Biotina/análogos & derivados , Bismuto/farmacocinética , Neoplasias Colorretais/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Radioisótopos de Chumbo/farmacocinética , Compostos Organometálicos/farmacocinética , Compostos Organometálicos/uso terapêutico , Radioimunoterapia/métodos , Animais , Biotina/farmacocinética , Biotina/uso terapêutico , Bismuto/uso terapêutico , Linhagem Celular Tumoral , Neoplasias Colorretais/radioterapia , Feminino , Humanos , Radioisótopos de Chumbo/uso terapêutico , Taxa de Depuração Metabólica , Camundongos , Camundongos Endogâmicos BALB C , Especificidade de Órgãos , Radioisótopos/farmacocinética , Radioisótopos/uso terapêutico , Compostos Radiofarmacêuticos/farmacocinética , Compostos Radiofarmacêuticos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
12.
Cancer Biother Radiopharm ; 20(1): 52-7, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15778581

RESUMO

INTRODUCTION: The somatostatin analog [DOTA(0)-Tyr(3)]-octreotide (DOTATOC) has been widely used to target somatostatin receptor expressing tumors for therapy using radionuclides such as (90)Y or (177)Lu. AIM: This aim of this study was to compare the effects of DOTATOC labeled to high linear energy transfer (LET) alpha-emitter (213)Bi and low-LET beta-emitter (177)Lu in vitro. MATERIALS AND METHODS: Somatostatin receptor (sstr)-positive cell line Capan-2 and sstr-negative control cell line A549 were used for the experiments. The effects of two exposure times using different radiation doses of high-LET alpha-emitter (213)Bi and low-LET beta-emitter (177)Lu were investigated using cell survival assay. The apoptotic effects were investigated using Cell Death Detection ELISA(PLUS)10x. The cumulated activity and the mean absorbed dose per unit cumulated activity were calculated using MIRD cellular Svalues. RESULTS: (213)Bi-DOTATOC had an approximately four times greater induction of apoptosis than (177)Lu-DOTATOC and a 100 times greater induction of apoptosis than nonradiolabeled DOTATOC. Nonspecific radiolabeled tetra-azacyclododecanetetra-acetic acid (DOTA) had a less pronounced effect on the cell survival and apoptosis, as compared to the sstr-specific radiolabeled DOTATOC. CONCLUSION: (213)Bi-DOTATOC is significantly more potent than (177)Lu-DOTATOC in vitro because of its high-LET alpha-emission.(213)Bi-DOTATOC shows enhanced effects on mitotic and apoptotic cell deaths.


Assuntos
Neoplasias/radioterapia , Octreotida/análogos & derivados , Receptores de Somatostatina/metabolismo , Actínio/uso terapêutico , Partículas alfa , Apoptose , Partículas beta , Bismuto/uso terapêutico , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular , Relação Dose-Resposta à Radiação , Transferência de Energia , Ensaio de Imunoadsorção Enzimática , Humanos , Ligantes , Lutécio/uso terapêutico , Mitose , Modelos Estatísticos , Octreotida/uso terapêutico , Radioisótopos/uso terapêutico , Radiometria , Compostos Radiofarmacêuticos/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...