Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Oncol ; 31(4): 729-40, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17786303

RESUMO

Despite their large size, antibodies have proven to be suitable radioisotope carriers to deliver systemic radiotherapy, often molecular image-based, for lymphoma and leukemia. To mimic antibody (Ab) targeting behavior while decreasing size by 50-100x, a combination of computational and experimental methods were used to generate molecules that bind to unique sites within the HLA-DR epitopic region of Lym-1, an Ab shown effective in patients. Lym-1 Ab mimics (synthetic high afinity ligands; SHALs) were generated and studied in vitro, using live cell binding assays, and/or pharmacokinetic studies over 24 h in xenografted mice given 1 or 20 microg SHAL doses i.v. Multimilligram amounts of each of the dimeric (bis) SHALs were synthesized at high purity, and labeled with indium-111 at high specific activity and purity. These SHALs were selective for HLA-DR and HLA-DR expressing malignant cells and had functional affinities that ranged from 10(-9) M (nanomolar) to 10(-10) M. Blood clearances ranged from 3.6 to 9.5 h and body clearances ranged from 15.2 to 43.0 h for the 6 bis DOTA-SHALs studied in a mouse model for non-Hodgkin's lymphoma (NHL). While localization was shown in Raji NHL xenografts, biodistribution was influenced by 'sinks' for individual ligands of the SHALs. Highly pure, dimeric mimics for HLA-DR Ab were synthesized, biotinylated and radiolabeled, and showed selectivity in vitro. Pharmacokinetic behavior in mice was influenced by the ligands and by the linker length of the dimeric SHALs. Nanomolar or better functional affinity was observed when a suitably long linker was used to connect the two bidentate SHALs. The concept and methodology are of interest because applicable for targeting most proteins; the SHAL synthetic platform is highly efficient and adaptive.


Assuntos
Anticorpos Monoclonais/metabolismo , Antígenos HLA-DR/imunologia , Linfoma não Hodgkin/diagnóstico por imagem , Mimetismo Molecular , Fragmentos de Peptídeos/imunologia , Compostos Radiofarmacêuticos/farmacocinética , Animais , Anticorpos Monoclonais Murinos , Ensaio de Imunoadsorção Enzimática , Feminino , Antígenos HLA-DR/metabolismo , Subtipos Sorológicos de HLA-DR , Humanos , Ligantes , Linfoma não Hodgkin/radioterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Estrutura Molecular , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/metabolismo , Radioimunoterapia , Cintilografia , Transplante Heterólogo
2.
Cancer Biother Radiopharm ; 22(4): 556-63, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17803451

RESUMO

UNLABELLED: Protease degradable linkers have been proposed to improve the therapeutic index (TI) (i.e., tumor to normal tissue) of molecular targeted radioisotope therapy by reducing unbound radiotargeting agent in the blood and other normal tissues. If the radioisotope is detached from the circulating targeting agent once the radioisotope level in the tumors has been maximized, the success of this system depends on the ability to anticipate a preferred intervention time that will lead to significantly improved TIs. This paper presents a method to predict preferred intervention times and TIs by using pharmacokinetic tracer studies carried out without intervention. METHODS: Pharmacokinetic data for the blood and tumors from tracer doses of 111In-labeled chimeric and mouse monoclonal antibodies in patients and in mice were used as surrogates for corresponding 90Y radioimmunoconjugates. Data were fit with simple pharmacokinetic functions. A set of formulas was then developed to estimate the improvement in therapeutic index and the preferred intervention time, using simple modeling assumptions. RESULTS: A modeled introduction of enzymatic cleavable linkers resulted in an increase in the tumor-to-blood TI by a factor of 3.2-1.6 for the systems analyzed. As expected, the preferred intervention times varied depending on the pharmacokinetic data, but could be predicted based on a priori knowledge of the actual or anticipated pharmacokinetics in the absence of intervention. CONCLUSIONS: These results highlight the potential value of cleavable linkers in substantially increasing the TI, and provide an approach for estimating a preferred intervention time, using actual or predicted pharmacokinetic data obtained without intervention.


Assuntos
Vasos Sanguíneos/enzimologia , Radioisótopos de Índio/metabolismo , Radioisótopos de Índio/farmacocinética , Peptídeos/metabolismo , Radioisótopos de Ítrio/metabolismo , Radioisótopos de Ítrio/farmacocinética , Animais , Linhagem Celular Tumoral , Humanos , Radioisótopos de Índio/química , Radioisótopos de Índio/uso terapêutico , Camundongos , Modelos Animais , Modelos Biológicos , Estudos Retrospectivos , Fatores de Tempo , Radioisótopos de Ítrio/química , Radioisótopos de Ítrio/uso terapêutico
3.
Int J Radiat Oncol Biol Phys ; 66(4): 1192-8, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16965873

RESUMO

PURPOSE: Radioactivity deposition in normal tissues limits the dose deliverable by radiopharmaceuticals (RP) in radioimmunotherapy (RIT). This study investigated the absorbed radiation dose in normal tissues for prostate cancer patients in comparison to breast cancer patients for 2 RPs using the monoclonal antibody (MAb) m170. METHODS AND MATERIALS: 111In-DOTA-glycylglycylglycyl-l-p-isothiocyanatophenylalanine amide (GGGF)-m170 and 111In-1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA) 2-iminothiolane (2IT)-m170, representing the same MAb and chelate with and without a cleavable linkage, were studied in 13 breast cancer and 26 prostate cancer patients. Dosimetry for 90Y was calculated using 111In MAb pharmacokinetics from the initial imaging study for each patient, using reference man- and patient-specific masses. RESULTS: The reference man-specific radiation doses (cGy/MBq) were not significantly different for the breast and the prostate cancer patients for both RPs in all but one tissue-RP combination (liver, DOTA-2IT). The patient-specific doses had differences between the groups most of which can be related to weight differences. CONCLUSIONS: Similar normal tissue doses were calculated for two groups of patients having different cancers and genders. This similarity combined with continued careful analysis of the imaging data might allow the use of higher starting doses in early phase RIT studies.


Assuntos
Anticorpos Monoclonais/farmacocinética , Neoplasias da Mama/metabolismo , Radioisótopos de Índio/farmacocinética , Neoplasias da Próstata/metabolismo , Radioisótopos de Ítrio/farmacocinética , Feminino , Humanos , Masculino , Taxa de Depuração Metabólica , Especificidade de Órgãos , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
4.
Clin Cancer Res ; 11(19 Pt 2): 7158s-7163s, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16203816

RESUMO

PURPOSE: Paclitaxel synergized radiolabeled monoclonal antibodies, enhancing therapeutic effect in studies in mice with human xenografts. Paclitaxel was also observed to increase tumor uptake in imaging studies of (111)In-DOTA-Gly3Phe-m170 in patients with breast and prostate cancers. Further evaluations of tissue-cumulated activities, therapeutic indices, and pharmacokinetics were done using data for patients with breast and prostate cancer and for mice with human breast cancer xenografts. EXPERIMENTAL DESIGN: In radioimmunotherapy trials, 12 patients with breast or prostate cancer were given two imaging doses (5 mCi each) of (111)In-DOTA-Gly3Phe-m170 1 week apart. Five of these patients were given a single dose of paclitaxel i.v. (75 mg/m2) 2 days after the second dose of (111)In. In a subsequent study, athymic mice with human breast cancer xenografts were given (111)In-DOTA-Gly3Phe-ChL6 alone, or in combination with daily paclitaxel i.p. (300 microg) one or more times. Pharmacokinetics were studied for at least 6 days in patients and 5 days in mice. Cumulated activities were determined for tumors and normal tissues. RESULTS: Tumor-cumulated activity for every patient in the paclitaxel-treated group increased for the second dose of (111)In-DOTA-Gly3Phe-m170. The median ratio of cumulated activities in tumors for imaging dose 2 to those for dose 1 was 1.0 (0.8-1.3) in patients that were not given paclitaxel and 1.3 (1.2-1.4) in patients given paclitaxel. Normal tissue-cumulated activities were not different for the two doses. Mice given paclitaxel 1 day after (111)In-DOTA-Gly3Phe-ChL6 also showed an increase in tumor-cumulated activity, 22.9 (+/- 1.3) versus 19.4 (+/- 3.3) microCi h/g/microCi (P = 0.05). Cumulated activities of normal tissues were similar for all groups of mice. CONCLUSIONS: Paclitaxel given 1 to 2 days after (111)In-DOTA-Gly3Phe-monoclonal antibody increased the tumor-cumulated activity in patients and in mice with epithelial cancers and did not alter cumulated activities in normal tissues.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Fitogênicos/farmacologia , Radioisótopos de Índio/uso terapêutico , Neoplasias Epiteliais e Glandulares/terapia , Paclitaxel/farmacologia , Radioimunoterapia/métodos , Animais , Anticorpos Monoclonais/química , Antineoplásicos Fitogênicos/administração & dosagem , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Compostos Heterocíclicos com 1 Anel/farmacologia , Humanos , Imunoglobulina G/química , Masculino , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Transplante de Neoplasias , Paclitaxel/administração & dosagem , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Fatores de Tempo
5.
Clin Cancer Res ; 11(19 Pt 2): 7187s-7194s, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16203820

RESUMO

PURPOSE: New strategies that target selected molecular characteristics and result in an effective therapeutic index are needed for metastatic, hormone-refractory prostate cancer. EXPERIMENTAL DESIGN: A series of preclinical and clinical studies were designed to increase the therapeutic index of targeted radiation therapy for prostate cancer. (111)In/90Y-monoclonal antibody (mAb), m170, which targets aberrant sugars on abnormal MUC1, was evaluated in androgen-independent prostate cancer patients to determine the maximum tolerated dose and efficacy of nonmyeloablative radioimmunotherapy and myeloablative combined modality radioimmunotherapy with paclitaxel. To enhance the tumor to liver therapeutic index, a cathepsin degradable mAb linkage ((111)In/90Y-peptide-m170) was used in the myeloablative combined modality radioimmunotherapy protocol. For tumor to marrow therapeutic index improvement in future studies, anti-MUC1 scFvs modules were developed for pretargeted radioimmunotherapy. Anti-MUC1 and anti-DOTA scFvs were conjugated to polyethylene glycol scaffolds tested on DU145 prostate cancer cells and prostate tissue arrays, along with mAbs against MUC1 epitopes. RESULTS: The nonmyeloablative maximum tolerated dose of 90Y-m170 was 0.74 GBq/m2 for patients with not more than 10% axial skeleton involvement. Metastatic prostate cancer was targeted in all 17 patients; mean radiation dose was 10.5 Gy/GBq and pain response occurred in 7 of 13 patients reporting pain. Myeloablative combined modality radioimmunotherapy with 0.4 GBq/m2 of 90Y-peptide-m170 and paclitaxel showed therapeutic effects in 4 of 6 patients and 30% less radiation to the liver per unit of activity. Neutropenia was dose limiting without marrow support and patient eligibility was a major limitation to dose escalation. Hypoglycosylated MUC1 epitopes were shown to be abundant in prostate cancer and to increase with disease grade. Anti-MUC1 scFvs binding to prostate cancer tissue and live cells were developed into di-scFv binding modules. CONCLUSIONS: The therapeutic index enhancement for prostate radioimmunotherapy was achieved in clinical studies by the addition of cathepsin cleavable linkers to 90Y-conjugated mAbs and the use of paclitaxel. However, the need for marrow support in myeloablative combined modality radioimmunotherapy restricted eligible patients. Therefore, modular pretargeted radioimmunotherapy, aiming at improving the tumor to marrow therapeutic index, is being developed.


Assuntos
Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Radioimunoterapia/métodos , Anticorpos Monoclonais , Antígenos/química , Antígenos de Neoplasias , Medula Óssea/metabolismo , Catepsinas/química , Terapia Combinada , Eletroforese em Gel de Poliacrilamida , Epitopos/química , Glicoproteínas/química , Humanos , Imuno-Histoquímica , Masculino , Maleimidas/química , Dose Máxima Tolerável , Modelos Químicos , Mucina-1 , Mucinas/química , Metástase Neoplásica , Paclitaxel/química , Biblioteca de Peptídeos , Peptídeos/química , Polietilenoglicóis/química , Ligação Proteica , Radiometria , Radioisótopos de Ítrio/química
6.
Clin Cancer Res ; 11(16): 5920-7, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16115934

RESUMO

PURPOSE: Although radioimmunotherapy alone is effective in lymphoma, its application to solid tumors will likely require a combined modality approach. In these phase I studies, paclitaxel was combined with radioimmunotherapy in patients with metastatic hormone-refractory prostate cancer or advanced breast cancer. EXPERIMENTAL DESIGN: Patients were imaged with indium-111 (111In)-1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid-peptide-m170. One week later, yttrium-90 (90Y)-m170 was infused (12 mCi/m2 for prostate cancer and 22 mCi/m2 for breast cancer). Initial cohorts received radioimmunotherapy alone. Subsequent cohorts received radioimmunotherapy followed 48 hours later by paclitaxel (75 mg/m2). Cyclosporine was given to prevent development of human anti-mouse antibody. RESULTS: Bone and soft tissue metastases were targeted by 111In-m170 in 15 of the 16 patients imaged. Three prostate cancer patients treated with radioimmunotherapy alone had no grade 3 or 4 toxicity. With radioimmunotherapy and paclitaxel, two of three prostate cancer patients developed transient grade 4 neutropenia. Four breast cancer patients treated with radioimmunotherapy alone had grade 3 or 4 myelosuppression. With radioimmunotherapy and paclitaxel, both breast cancer patients developed grade 4 neutropenia. Three breast cancer patients required infusion of previously harvested peripheral blood stem cells because of neutropenic fever or bleeding. One patient in this trial developed human anti-mouse antibody in contrast to 12 of 17 patients in a prior trial using m170-radioimmunotherapy without cyclosporine. CONCLUSIONS: 111In/90Y-m170 targets prostate and breast cancer and can be combined with paclitaxel with toxicity limited to marrow suppression at the dose levels above. The maximum tolerated dose of radioimmunotherapy and fixed-dose paclitaxel with peripheral blood stem cell support has not been reached. Cyclosporine is effective in preventing human anti-mouse antibody, suggesting the feasibility of multidose, "fractionated" therapy that could enhance clinical response.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Paclitaxel/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/radioterapia , Radioimunoterapia/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Fitogênicos/efeitos adversos , Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias Ósseas/prevenção & controle , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Catepsinas/metabolismo , Terapia Combinada , Ciclosporina/metabolismo , Ciclosporina/uso terapêutico , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Feminino , Compostos Heterocíclicos com 1 Anel/química , Compostos Heterocíclicos com 1 Anel/farmacocinética , Humanos , Radioisótopos de Índio/química , Radioisótopos de Índio/farmacocinética , Radioisótopos de Índio/uso terapêutico , Masculino , Camundongos , Pessoa de Meia-Idade , Neutropenia/etiologia , Paclitaxel/efeitos adversos , Neoplasias da Próstata/patologia , Radioimunoterapia/efeitos adversos , Neoplasias de Tecidos Moles/prevenção & controle , Neoplasias de Tecidos Moles/secundário , Resultado do Tratamento , Radioisótopos de Ítrio/química , Radioisótopos de Ítrio/farmacocinética , Radioisótopos de Ítrio/uso terapêutico
7.
J Nucl Med ; 46(6): 1034-41, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15937317

RESUMO

UNLABELLED: Myelotoxicity can be ameliorated by peripheral blood stem cell (PBSC) infusion. Continuous irradiation by radioactivity retained in the body after high-dose radioimmunotherapy can damage PBSCs if they are transfused too early. Previously, infusion time was predetermined using the radioactivity concentration in the blood. This study proposes to plan PBSC infusion time based on noninvasive dosimetry that considers damage of PBSCs during PBSC circulation and residence in organs with high radioactivity. METHODS: The method considers a time-varying distribution of PBSCs and radioactivity in tissues. Five breast cancer patients received (111)In-2IT-BAD-m170 for imaging, and 3 of the 5 received high doses of (90)Y-2IT-BAD-m170 therapy followed by PBSC infusion. (90)Y concentrations in tissues were extrapolated from quantitative imaging of (111)In, and (90)Y blood concentrations were determined from (90)Y in serial blood samples. The radiation dose to PBSCs was determined by time integration of the organ dose rate and PBSC distribution rate. The radiosensitivity of PBSCs was determined by measuring survival of granulocyte-macrophage colony-forming units with (90)Y in cell culture. RESULTS: The mean effective half-life of (90)Y within the imaging period (up to 6 d) was 3.7 d for liver, 2.4 d for spleen, 2.1 d for kidneys, 1.8 d for lungs, and 1.6 d for blood. The survival fractions of PBSCs in patients were determined as functions of the infusion time and the injected dose of (90)Y-2IT-BAD-m170. To achieve 90% PBSC survival rate for a 2.0-GBq injection dose, PBSC dosimetry suggested a time interval of 13 d after radioimmunotherapy for PBSC infusion. In contrast, the simple blood concentration method suggested an interval about 7 d for the same PBSC survival rate. In our clinical practice, an interval of 2 wk has been used and worked well. CONCLUSION: A noninvasive dosimetry method was developed for optimizing the time interval for PBSC infusion after high-dose radionuclide therapy. Our studies suggested that the PBSC dosimetry method was more effective than the blood concentration method in determining the optimal time to reinfuse PBSCs for radiopharmaceuticals that have much a higher activity concentration in organs than that in the blood.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias da Mama/radioterapia , Transplante de Células-Tronco de Sangue Periférico , Radioimunoterapia , Compostos Radiofarmacêuticos/uso terapêutico , Antígenos Glicosídicos Associados a Tumores/imunologia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/imunologia , Células Cultivadas , Feminino , Humanos , Radioisótopos de Índio , Radiometria , Cintilografia , Fatores de Tempo , Radioisótopos de Ítrio/uso terapêutico
8.
Cancer Biother Radiopharm ; 20(6): 662-70, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16398618

RESUMO

UNLABELLED: Splenomegaly is frequently found in non-Hodgkin's lymphoma (NHL) patients. This study evaluated the implications of splenic volume change in response to radioimmunotherapy (RIT) using radiolabeled Lym- 1 antibody. METHODS: Twenty-nine NHL patients treated with radiolabeled-Lym-1 and 9 breast cancer patients, the reference group, treated with radiolabeled ChL6, BrE-3, or m170, were analyzed using X-ray computer tomography (CT) splenic images obtained before and after RIT. Patient-specific radiation doses to the spleen were determined using actual splenic volume determined by CT and body weight. RESULTS: Of 29 NHL patients, 13 that had splenic volumes equal or less than 310 mL, there was little or no change in splenic volume after RIT, despite splenic radiation doses as high as 23.1 Gy (median 8.0 Gy). Similarly, in a reference group of 9 breast cancer patients, there was little or no change in splenic volume after RIT, despite doses as high as 14.4 Gy (median 11.5 Gy). In the remaining 16 NHL patients, splenic volumes decreased in 13 patients, with initial volumes of 380-1,400 mL, by 68-548 mL despite splenic radiation doses as low as 1.1 Gy (median 3.2 Gy); splenic volumes increased in the other 3 patients after RIT. Although not statistically significant in this small series, therapeutic remission, defined conventionally by nodal tumor response, was more likely when splenic volume decreased after RIT. All 10 NHL patients with greater than a 15% decrease in their splenic volumes after RIT had nodal tumor response (5 complete response, 5 partial response). There were 12 responders (5 complete response and 7 partial response) in 19 NHL patients with less than a 15% decrease in splenic volume after RIT. CONCLUSIONS: Splenic volume decreased in NHL patients with splenomegaly, despite splenic radiation dose as low as 1.1 Gy. In the absence of splenomegaly, splenic volume did not decrease, even after much higher radiation doses. RIT with radiolabeled-Lym-1 may benefit NHL patients with splenomegaly, with reduction in splenic volume likely owing to a therapeutic effect on malignant lymphocytes.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias da Mama/radioterapia , Radioisótopos de Índio/uso terapêutico , Radioisótopos do Iodo/uso terapêutico , Linfoma não Hodgkin/radioterapia , Radioimunoterapia/efeitos adversos , Baço/anatomia & histologia , Esplenomegalia/diagnóstico por imagem , Anticorpos Monoclonais Murinos , Feminino , Humanos , Linfonodos/efeitos da radiação , Cintilografia , Estudos Retrospectivos , Baço/efeitos da radiação , Resultado do Tratamento
9.
Cancer Biother Radiopharm ; 18(2): 231-7, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12804049

RESUMO

Radionuclides provide biologically-distributed vehicles for radiotherapy of multifocal cancer. Two algorithms, fixed vs individualized, have been used to prescribe the therapeutic dose of radionuclide (GBq) for the patient. The individualized method for prescribing radionuclide dose takes variations in drug pharmacokinetics into consideration, whereas the fixed method depends, in part, on documentation that there is little interpatient pharmacokinetic variability for the radiolabeled drug. Two data bases, selected to compare iodine-131((131)I) and indium-111((111)In) labeled MAbs, were used to assess interpatient pharmacokinetic variability and its impact on radionuclide dose prescription. Pharmacokinetic data obtained over 7 days for non-Hodgkins lymphoma (NHL) patients given (131)I-Lym-1 (n = 46) or (111)In-Lym-1 (n = 13) were used to obtain cumulated activities. Although (131)I-Lym-1 often showed greater interpatient variability, (111)In-Lym-1 showed several-fold variability for many tissues. Both (131)I- and (111)In-Lym-1 had sufficient interpatient variability to be significant for radionuclide dose prescription, depending on the dose-limiting critical tissue. Interpatient variability exceeded intra- and interoperator variability and intrapatient variability over time for a single institution. In summary, the magnitude of interpatient pharmacokinetic variability for (131)I- and (111)In-Lym-1 suggested that an optimally safe and effective therapy can be best achieved when radionuclide dose is influenced by estimated radiation dose, if the latter is reproducible from institution to institution.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Radioisótopos de Índio/uso terapêutico , Linfoma não Hodgkin/radioterapia , Radioimunoterapia , Projetos de Pesquisa , Radioisótopos de Ítrio/uso terapêutico , Adulto , Idoso , Algoritmos , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Murinos , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Radioisótopos de Índio/farmacocinética , Masculino , Pessoa de Meia-Idade , Dosagem Radioterapêutica , Distribuição Tecidual , Radioisótopos de Ítrio/farmacocinética
10.
Cancer Biother Radiopharm ; 18(1): 71-9, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12667310

RESUMO

Recent clinical results have demonstrated the promise of targeted radionuclide therapy for advanced cancer. As the success of this emerging form of radiation therapy grows, accurate treatment planning and radiation dose simulations are likely to become increasingly important. To address this need, we have initiated the development of a new, Monte Carlo transport-based treatment planning system for molecular targeted radiation therapy as part of the MINERVA system. The goal of the MINERVA dose calculation system is to provide 3-D Monte Carlo simulation-based dosimetry for radiation therapy, focusing on experimental and emerging applications. For molecular targeted radionuclide therapy applications, MINERVA calculates patient-specific radiation dose estimates using computed tomography to describe the patient anatomy, combined with a user-defined 3-D radiation source. This paper describes the validation of the 3-D Monte Carlo transport methods to be used in MINERVA for molecular targeted radionuclide dosimetry. It reports comparisons of MINERVA dose simulations with published absorbed fraction data for distributed, monoenergetic photon and electron sources, and for radioisotope photon emission. MINERVA simulations are generally within 2% of EGS4 results and 10% of MCNP results, but differ by up to 40% from the recommendations given in MIRD Pamphlets 3 and 8 for identical medium composition and density. For several representative source and target organs in the abdomen and thorax, specific absorbed fractions calculated with the MINERVA system are generally within 5% of those published in the revised MIRD Pamphlet 5 for 100 keV photons. However, results differ by up to 23% for the adrenal glands, the smallest of our target organs. Finally, we show examples of Monte Carlo simulations in a patient-like geometry for a source of uniform activity located in the kidney.


Assuntos
Método de Monte Carlo , Neoplasias/radioterapia , Radioisótopos/uso terapêutico , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador , Algoritmos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA