Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
1.
Phys Chem Chem Phys ; 26(17): 12915-12927, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38629229

RESUMO

Targeted alpha therapy (TAT) is a methodology that is being developed as a promising cancer treatment using the α-particle decay of radionuclides. This technique involves the use of heavy radioactive elements being placed near the cancer target area to cause maximum damage to the cancer cells while minimizing the damage to healthy cells. Using gold nanoparticles (AuNPs) as carriers, a more effective therapy methodology may be realized. AuNPs can be good candidates for transporting these radionuclides to the vicinity of the cancer cells since they can be labeled not just with the radionuclides, but also a host of other proteins and ligands to target these cells and serve as additional treatment options. Research has shown that astatine and iodine are capable of adsorbing onto the surface of gold, creating a covalent bond that is quite stable for use in experiments. However, there are still many challenges that lie ahead in this area, whether they be theoretical, experimental, and even in real-life applications. This review will cover some of the major developments, as well as the current state of technology, and the problems that need to be tackled as this research topic moves along to maturity. The hope is that with more workers joining the field, we can make a positive impact on society, in addition to bringing improvement and more knowledge to science.


Assuntos
Astato , Ouro , Nanopartículas Metálicas , Ouro/química , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Humanos , Astato/química , Astato/uso terapêutico , Neoplasias/radioterapia , Neoplasias/tratamento farmacológico
2.
Nucl Med Biol ; 126-127: 108387, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37837782

RESUMO

The alpha emitter astatine-211 (211At) is a promising candidate for cancer treatment based on Targeted Alpha (α) Therapy (TAT). A small number of facilities, distributed across the United States, are capable of accelerating α-particle beams to produce 211At. However, challenges remain regarding strategic methods for shipping 211At in a form adaptable to advanced radiochemistry reactions and other uses of the radioisotope. PURPOSE: Our method allows shipment of 211At in various quantities in a form convenient for further radiochemistry. PROCEDURES: For this study, a 3-octanone impregnated Amberchrom CG300M resin bed in a column cartridge was used to separate 211At from the bismuth matrix on site at the production accelerator (Texas A&M) in preparation for shipping. Aliquots of 6 M HNO3 containing up to ≈2.22 GBq of 211At from the dissolved target were successfully loaded and retained on columns. Exempt packages (<370 MBq) were shipped to a destination radiochemistry facility, University of Texas MD Anderson Cancer Center, in the form of a convenient air-dried column. Type A packages have been shipped overnight to University of Alabama at Birmingham. MAIN FINDINGS: Air-dried column hold times of various lengths did not inhibit simple and efficient recovery of 211At. Solution eluted from the column was sufficiently high in specific activity to successfully radiolabel a model compound, 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (1), with 211At. The method to prepare and ship 211At described in this manuscript has also been used to ship larger quantities of 211At a greater distance to University of Alabama at Birmingham. PRINCIPAL CONCLUSIONS: The successful proof of this method paves the way for the distribution of 211At from Texas A&M University to research institutions and clinical oncology centers in Texas and elsewhere. Use of this simple method at other facilities has the potential increase the overall availability of 211At for preclinical and clinical studies.


Assuntos
Astato , Humanos , Astato/uso terapêutico , Astato/química , Radioisótopos/química , Partículas alfa/uso terapêutico , Radioquímica/métodos
3.
Eur J Nucl Med Mol Imaging ; 50(3): 849-858, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36344651

RESUMO

PURPOSE: Targeted α-therapy (TAT) for prostate-specific membrane antigen (PSMA) is a promising treatment for metastatic castration-resistant prostate cancer (CRPC). Astatine is an α-emitter (half-life=7.2 h) that can be produced by a 30-MeV cyclotron. This study evaluated the treatment effect of 211At-labeled PSMA compounds in mouse xenograft models. METHODS: Tumor xenograft models were established by subcutaneous transplantation of human prostate cancer cells (LNCaP) in NOD/SCID mouse. [211At]PSMA1, [211At]PSMA5, or [211At]PSMA6 was administered to LNCaP xenograft mice to evaluate biodistribution at 3 and 24 h. The treatment effect was evaluated by administering [211At]PSMA1 (0.40 ± 0.07 MBq), [211At]PSMA5 (0.39 ± 0.03 MBq), or saline. Histopathological evaluation was performed for the at-risk organs at 3 and 6 weeks after administration. RESULTS: [211At]PSMA5 resulted in higher tumor retention compared to [211At]PSMA1 and [211At]PSMA6 (30.6 ± 17.8, 12.4 ± 4.8, and 19.1 ± 4.5 %ID/g at 3 h versus 40.7 ± 2.6, 8.7 ± 3.5, and 18.1 ± 2.2%ID/g at 24 h, respectively), whereas kidney excretion was superior in [211At]PSMA1 compared to [211At]PSMA5 and [211At]PSMA6. An excellent treatment effect on tumor growth was observed after [211At]PSMA5 administration. [211At]PSMA1 also showed a substantial treatment effect; however, the tumor size was relatively larger compared to that with [211At]PSMA5. In the histopathological evaluation, regenerated tubules were detected in the kidneys at 3 and 6 weeks after the administration of [211At]PSMA5. CONCLUSION: TAT using [211At]PSMA5 resulted in excellent tumor growth suppression with minimal side effects in the normal organs. [211At]PSMA5 should be considered a new possible TAT for metastatic CRPC, and translational prospective trials are warranted.


Assuntos
Astato , Neoplasias de Próstata Resistentes à Castração , Neoplasias da Próstata , Masculino , Humanos , Animais , Camundongos , Astato/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/diagnóstico por imagem , Neoplasias de Próstata Resistentes à Castração/radioterapia , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Distribuição Tecidual , Estudos Prospectivos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias da Próstata/patologia , Glutamato Carboxipeptidase II/metabolismo , Antígenos de Superfície/metabolismo , Linhagem Celular Tumoral , Compostos Radiofarmacêuticos/uso terapêutico
4.
Gan To Kagaku Ryoho ; 49(8): 829-834, 2022 Aug.
Artigo em Japonês | MEDLINE | ID: mdl-36046965

RESUMO

Radioactive iodine has long been used clinically for patients with differentiated thyroid cancer. Radioiodine(131I) is used for the ablation of thyroid remnants or treatment of metastatic thyroid cancer. However, some patients with multiple metastases are refractory to repetitive 131I treatment, despite the targeted regions showing sufficient iodine uptake. In such patients, ß- particle therapy using 131I is inadequate and another strategy is needed using more effective radionuclide targeting the sodium/iodide symporter(NIS). Astatine(211At)is receiving increasing attention as an α-emitter for targeted radionuclide therapy. 211At is a halogen element with similar chemical properties to iodine. α particles emitted from 211At has higher linear energy transfer as compared to ß particles from 131I and exert a better therapeutic effect by inducing DNA double strand breaks and free radical formation. We showed that increase of the radiochemical purity of astatide of 211At solution by addition of ascorbic acid was associated with significantly enhanced uptake of 211At by both normal thyroid tissue and differentiated thyroid cancer cells. The treatment effect of 211At solution in the K1-NIS xenograft model was dose-dependent and was associated with prolonged survival, suggesting the potential applicability of targeted α therapy for the treatment of advanced differentiated thyroid cancer. Thus, targeted α therapy using 211At is highly promising for the treatment of advanced differentiated thyroid cancer. We have already started the clinical trial of 211At-NaAt in Osaka University Hospital since November 2021 after getting the approval by IRB and PMDA investigation. We would like to get the proof of concept that astatine can be used safely and effectively in patients, aiming at the drug approval as a targeted α therapeutic from Japan.


Assuntos
Adenocarcinoma , Astato , Neoplasias da Glândula Tireoide , Adenocarcinoma/tratamento farmacológico , Astato/uso terapêutico , Ensaios Clínicos como Assunto , Humanos , Radioisótopos do Iodo/uso terapêutico , Neoplasias da Glândula Tireoide/tratamento farmacológico
5.
Int J Mol Sci ; 23(16)2022 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-36012698

RESUMO

Astatine (211At) is an alpha-emitter with a better treatment efficacy against differentiated thyroid cancer compared with iodine (131I), a conventional beta-emitter. However, its therapeutic comparison has not been fully evaluated. In this study, we compared the therapeutic effect between [211At]NaAt and [131I]NaI. In vitro analysis of a double-stranded DNA break (DSB) and colony formation assay were performed using K1-NIS cells. The therapeutic effect was compared using K1-NIS xenograft mice administered with [211At]NaAt (0.4 MBq (n = 7), 0.8 MBq (n = 9), and 1.2 MBq (n = 4)), and [131I]NaI (1 MBq (n = 4), 3 MBq (n = 4), and 8 MBq (n = 4)). The [211At]NaAt induced higher numbers of DSBs and had a more reduced colony formation than [131I]NaI. In K1-NIS mice, dose-dependent therapeutic effects were observed in both [211At]NaAt and [131I]NaI. In [211At]NaAt, a stronger tumour-growth suppression was observed, while tumour regrowth was not observed until 18, 25, and 46 days after injection of 0.4, 0.8, and 1.2 MBq of [211At]NaAt, respectively. While in [131I]NaI, this was observed within 12 days after injection (1, 3, and 8 MBq). The superior therapeutic effect of [211At]NaAt suggests the promising clinical applicability of targeted alpha therapy using [211At]NaAt in patients with differentiated thyroid cancer refractory to standard [131I]NaI treatment.


Assuntos
Adenocarcinoma , Astato , Neoplasias da Glândula Tireoide , Adenocarcinoma/tratamento farmacológico , Animais , Astato/uso terapêutico , Humanos , Radioisótopos do Iodo/uso terapêutico , Camundongos , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/genética , Transplante Heterólogo
6.
Clin Cancer Res ; 28(18): 4146-4157, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35861867

RESUMO

PURPOSE: [131I]meta-iodobenzylguanidine ([131I]MIBG) is a targeted radiotherapeutic administered systemically to deliver beta particle radiation in neuroblastoma. However, relapses in the bone marrow are common. [211At]meta-astatobenzylguanidine ([211At] MABG) is an alpha particle emitter with higher biological effectiveness and short path length which effectively sterilizes microscopic residual disease. Here we investigated the safety and antitumor activity [211At]MABG in preclinical models of neuroblastoma. EXPERIMENTAL DESIGN: We defined the maximum tolerated dose (MTD), biodistribution, and toxicity of [211At]MABG in immunodeficient mice in comparison with [131I]MIBG. We compared the antitumor efficacy of [211At]MABG with [131I]MIBG in three murine xenograft models. Finally, we explored the efficacy of [211At]MABG after tail vein xenografting designed to model disseminated neuroblastoma. RESULTS: The MTD of [211At]MABG was 66.7 MBq/kg (1.8 mCi/kg) in CB17SC scid-/- mice and 51.8 MBq/kg (1.4 mCi/kg) in NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice. Biodistribution of [211At]MABG was similar to [131I]MIBG. Long-term toxicity studies on mice administered with doses up to 41.5 MBq/kg (1.12 mCi/kg) showed the radiotherapeutic to be well tolerated. Both 66.7 MBq/kg (1.8 mCi/kg) single dose and fractionated dosing 16.6 MBq/kg/fraction (0.45 mCi/kg) × 4 over 11 days induced marked tumor regression in two of the three models studied. Survival was significantly prolonged for mice treated with 12.9 MBq/kg/fraction (0.35 mCi/kg) × 4 doses over 11 days [211At]MABG in the disseminated disease (IMR-05NET/GFP/LUC) model (P = 0.003) suggesting eradication of microscopic disease. CONCLUSIONS: [211At]MABG has significant survival advantage in disseminated models of neuroblastoma. An alpha particle emitting radiopharmaceutical may be effective against microscopic disseminated disease, warranting clinical development.


Assuntos
Astato , Neuroblastoma , 3-Iodobenzilguanidina/efeitos adversos , Partículas alfa/uso terapêutico , Animais , Astato/uso terapêutico , Guanidinas/uso terapêutico , Humanos , Radioisótopos do Iodo/uso terapêutico , Camundongos , Camundongos Endogâmicos NOD , Recidiva Local de Neoplasia/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Neuroblastoma/radioterapia , Compostos Radiofarmacêuticos/efeitos adversos , Distribuição Tecidual , Células Tumorais Cultivadas
7.
Leukemia ; 36(6): 1485-1491, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35474099

RESUMO

Radioimmunotherapy (RIT) has long been pursued to improve outcomes in acute leukemia and higher-risk myelodysplastic syndrome (MDS). Of increasing interest are alpha-particle-emitting radionuclides such as astatine-211 (211At) as they deliver large amounts of radiation over just a few cell diameters, enabling efficient and selective target cell kill. Here, we developed 211At-based RIT targeting CD123, an antigen widely displayed on acute leukemia and MDS cells including underlying neoplastic stem cells. We generated and characterized new murine monoclonal antibodies (mAbs) specific for human CD123 and selected four, all of which were internalized by CD123+ target cells, for further characterization. All mAbs could be conjugated to a boron cage, isothiocyanatophenethyl-ureido-closo-decaborate(2-) (B10), and labeled with 211At. CD123+ cell targeting studies in immunodeficient mice demonstrated specific uptake of 211At-labeled anti-CD123 mAbs in human CD123+ MOLM-13 cell tumors in the flank. In mice injected intravenously with MOLM-13 cells or a CD123NULL MOLM-13 subline, a single dose of up to 40 µCi of 211At delivered via anti-CD123 mAb decreased tumor burdens and substantially prolonged survival dose dependently in mice bearing CD123+ but not CD123- leukemia xenografts, demonstrating potent and target-specific in vivo anti-leukemia efficacy. These data support the further development of 211At-CD123 RIT toward clinical application.


Assuntos
Astato , Leucemia Mieloide Aguda , Doença Aguda , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Astato/uso terapêutico , Humanos , Subunidade alfa de Receptor de Interleucina-3 , Leucemia Mieloide Aguda/tratamento farmacológico , Camundongos , Radioimunoterapia
8.
J Nanobiotechnology ; 19(1): 223, 2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34320997

RESUMO

BACKGROUND: 211At is a high-energy α-ray emitter with a relatively short half-life and a high cytotoxicity for cancer cells. Its dispersion can be imaged using clinical scanners, and it can be produced in cyclotrons without the use of nuclear fuel material. This study investigated the biodistribution and the antitumor effect of 211At-labeled gold nanoparticles (211At-AuNP) administered intratumorally. RESULTS: AuNP with a diameter of 5, 13, 30, or 120 nm that had been modified with poly (ethylene glycol) methyl ether (mPEG) thiol and labeled with 211At (211At-AuNP-S-mPEG) were incubated with tumor cells, or intratumorally administered to C6 glioma or PANC-1 pancreatic cancers subcutaneously transplanted into rodent models. Systemic and intratumoral distributions of the particles in the rodents were then evaluated using scintigraphy and autoradiography, and the changes in tumor volumes were followed for about 40 days. 211At-AuNP-S-mPEG was cytotoxic when it was internalized by the tumor cells. After intratumoral administration, 211At-AuNP-S-mPEG became localized in the tumor and did not spread to systemic organs during a time period equivalent to 6 half-lives of 211At. Tumor growth was strongly suppressed for both C6 and PANC-1 by 211At-AuNP-S-mPEG. In the C6 glioma model, the strongest antitumor effect was observed in the group treated with 211At-AuNP-S-mPEG with a diameter of 5 nm. CONCLUSIONS: The intratumoral single administration of a simple nanoparticle, 211At-AuNP-S-mPEG, was shown to suppress the growth of tumor tissue strongly in a particle size-dependent manner without radiation exposure to other organs caused by systemic spread of the radionuclide.


Assuntos
Astato/uso terapêutico , Ouro/uso terapêutico , Nanopartículas/química , Nanopartículas/uso terapêutico , Coloração e Rotulagem/métodos , Animais , Astato/química , Glioma , Ouro/química , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Tamanho da Partícula , Polietilenoglicóis , Cintilografia/métodos , Ratos , Distribuição Tecidual
9.
Nucl Med Biol ; 94-95: 67-80, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33601187

RESUMO

INTRODUCTION: The high potency and short tissue range of α-particles are attractive features for targeted radionuclide therapy, particularly for cancers with micro-metastases. In the current study, we describe the synthesis of a series of 211At-labeled prostate-specific membrane antigen (PSMA) inhibitors and their preliminary evaluation as potential agents for metastatic prostate cancer treatment. METHODS: Four novel Glu-urea based PSMA ligands containing a trialkyl stannyl group were synthesized and labeled with 211At, and for comparative purposes, 131I, via halodestannylation reactions with N-chlorosuccinimide as the oxidant. A PSMA inhibitory assay was performed to evaluate PSMA binding of the unlabeled, iodinated compounds. A series of paired-label biodistribution experiments were performed to compare each 211At-labeled PSMA ligand to its 131I-labeled counterpart in mice bearing subcutaneous PC3 PSMA+ PIP xenografts. RESULTS: Radiochemical yields ranged from 32% to 65% for the 211At-labeled PSMA inhibitors and were consistently lower than those obtained with the corresponding 131I-labeled analogue. Good localization in PC3 PSMA+ PIP but not control xenografts was observed for all labeled molecules studied, which exhibited a variable degree of in vivo dehalogenation as reflected by thyroid and stomach activity levels. Normal tissue uptake and in vivo stability for several of the compounds was markedly improved compared with the previously evaluated compounds, [211At]DCABzL and [*I]DCIBzL. CONCLUSIONS AND IMPLICATIONS FOR PATIENT CARE: Compared with the first generation compound [211At]DCABzL, several of the novel 211At-labeled PSMA ligands exhibited markedly improved stability in vivo and higher tumor-to-normal tissue ratios. [211At]GV-620 has the most promising characteristics and warrants further evaluation as a targeted radiotherapeutic for prostate cancer.


Assuntos
Partículas alfa/uso terapêutico , Antígenos de Superfície/metabolismo , Astato/uso terapêutico , Glutamato Carboxipeptidase II/metabolismo , Neoplasias da Próstata/radioterapia , Animais , Linhagem Celular Tumoral , Humanos , Ligantes , Masculino , Camundongos , Neoplasias da Próstata/patologia , Distribuição Tecidual
10.
Cancer Sci ; 112(5): 1975-1986, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33606344

RESUMO

Tissue factor (TF), the trigger protein of the extrinsic blood coagulation cascade, is abundantly expressed in various cancers including gastric cancer. Anti-TF monoclonal antibodies (mAbs) capable of targeting cancers have been successfully applied to armed antibodies such as antibody-drug conjugates (ADCs) and molecular imaging probes. We prepared an anti-TF mAb, clone 1084, labeled with astatine-211 (211 At), as a promising alpha emitter for cancer treatment. Alpha particles are characterized by high linear energy transfer and a range of 50-100 µm in tissue. Therefore, selective and efficient tumor accumulation of alpha emitters results in potent antitumor activities against cancer cells with minor effects on normal cells adjacent to the tumor. Although the 211 At-conjugated clone 1084 (211 At-anti-TF mAb) was disrupted by an 211 At-induced radiochemical reaction, we demonstrated that astatinated anti-TF mAbs eluted in 0.6% or 1.2% sodium ascorbate (SA) solution were protected from antibody denaturation, which contributed to the maintenance of cellular binding activities and cytocidal effects of this immunoconjugate. Although body weight loss was observed in mice administered a 1.2% SA solution, the loss was transient and the radioprotectant seemed to be tolerable in vivo. In a high TF-expressing gastric cancer xenograft model, 211 At-anti-TF mAb in 1.2% SA exerted a significantly greater antitumor effect than nonprotected 211 At-anti-TF mAb. Moreover, the antitumor activities of the protected immunoconjugate in gastric cancer xenograft models were dependent on the level of TF in cancer cells. These findings suggest the clinical availability of the radioprotectant and applicability of clone 1084 to 211 At-radioimmunotherapy.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Ácido Ascórbico/uso terapêutico , Astato/uso terapêutico , Imunoconjugados/uso terapêutico , Radioimunoterapia/métodos , Neoplasias Gástricas/terapia , Tromboplastina/imunologia , Animais , Anticorpos Monoclonais Humanizados/farmacocinética , Astato/farmacocinética , Coagulação Sanguínea/fisiologia , Peso Corporal , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , Imunoconjugados/química , Imunoconjugados/farmacocinética , Transferência Linear de Energia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Desnaturação Proteica , Protetores contra Radiação/uso terapêutico , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Tromboplastina/metabolismo
11.
Nucl Med Biol ; 90-91: 15-22, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32916470

RESUMO

INTRODUCTION: Targeted α-radionuclide therapy has attracted attention as a promising therapy for refractory cancers. However, the application is limited to certain types of cancer. Since L-type amino acid transporter 1 (LAT1) is highly expressed in various human cancers, we prepared an LAT1-selective α-radionuclide-labeled amino acid analog, 2-[211At]astato-α-methyl-L-phenylalanine (2-[211At]AAMP), and evaluated its potential as a therapeutic agent. METHODS: 2-[211At]AAMP was prepared from the stannyl precursor. Stability of 2-[211At]AAMP was evaluated both in vitro and in vivo. In vitro studies using an LAT1-expressing human ovarian cancer cell line, SKOV3, were performed to evaluate cellular uptake and cytotoxicity of 2-[211At]AAMP. Biodistribution and therapeutic studies in SKOV3-bearing mice were performed after intravenous injection of 2-[211At]AAMP. RESULTS: 2-[211At]AAMP was stable in murine plasma in vitro and excreted intact into urine. Cellular uptake of 2-[211At]AAMP was inhibited by treatment with an LAT1-selective inhibitor. After 24 h incubation, 2-[211At]AAMP suppressed clonogenic growth at 10 kBq/ml, and induced cell death and DNA double-strand breaks at 25 kBq/ml. When injected into mice, 2-[211At]AAMP exhibited peak accumulation in the tumor at 30 min postinjection, and radioactivity levels in the tumor were retained up to 60 min. The majority of the radioactivity was rapidly eliminated from the body into urine in an intact form immediately after injection. 2-[211At]AAMP significantly improved the survival of mice (P < 0.05) without serious side effects. CONCLUSION: 2-[211At]AAMP showed α-radiation-dependent cellular growth inhibition after it was taken up via LAT1. In addition, 2-[211At]AAMP had a beneficial effect on survival in vivo. These findings suggest that 2-[211At]AAMP would be useful for the treatment of LAT1-positive cancer. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: This is the first report of an LAT1-targeting radiopharmaceutical for α-radionuclide therapy; this agent would be applicable for the treatment of various types of cancer.


Assuntos
Astato/uso terapêutico , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Fenilalanina/química , Fenilalanina/uso terapêutico , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Camundongos , Fenilalanina/farmacocinética , Tomografia por Emissão de Pósitrons , Distribuição Tecidual
12.
Sci Rep ; 10(1): 6810, 2020 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-32321944

RESUMO

To explore stem-cell-targeted radioimmunotherapy with α-particles in acute myelogenous leukemia (AML), pharmacokinetics and dosimetry of the 211At-labeled anti-C-X-C chemokine receptor type 4 monoclonal antibody (211At-CXCR4 mAb) were conducted using tumor xenografted mice. The biological half-life of 211At-CXCR4 mAb in blood was 15.0 h. The highest tumor uptake of 5.05%ID/g with the highest tumor-to-muscle ratio of 8.51 ± 6.14 was obtained at 6 h. Radiation dosimetry estimated with a human phantom showed absorbed doses of 0.512 mGy/MBq in the bone marrow, 0.287 mGy/MBq in the kidney, and <1 mGy/MBq in other major organs except bone. Sphere model analysis revealed 22.8 mGy/MBq in a tumor of 10 g; in this case, the tumor-to-bone marrow and tumor-to-kidney ratios were 44.5 and 79.4, respectively. The stem-cell-targeted α-particle therapy using 211At-CXCR4 mAb for AML appears possible and requires further therapeutic studies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Astato/uso terapêutico , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/terapia , Células-Tronco Neoplásicas/patologia , Radioimunoterapia , Receptores CXCR4/imunologia , Animais , Humanos , Radioisótopos do Iodo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Músculos/patologia , Especificidade de Órgãos , Doses de Radiação , Distribuição Tecidual , Células U937 , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Biother Radiopharm ; 35(7): 511-519, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32109139

RESUMO

Background: In a previous clinical study, the authors evaluated the potential of antitenascin C monoclonal antibody (mAb) 81C6 labeled with 211At via the prosthetic agent N-succinimidyl 3-[211At]astatobenzoate (SAB) for the treatment of primary brain tumors. Although encouraging results were obtained, labeling chemistry failed while attempting to escalate the dose to 370 MBq. The goal of the current study was to develop a revised procedure less susceptible to radiolysis-mediated effects on 211At labeling that would be suitable for use at higher activity levels of this α-emitter. Materials and Methods: Addition of N-chlorosuccinimide to the methanol used to remove the 211At from the cryotrap after bismuth target distillation was done to thwart radiolytic decomposition of reactive 211At and the tin precursor. A series of 11 reactions were performed to produce SAB at initial 211At activity levels of 0.31-2.74 GBq from 50 µg of N-succinimidyl 3-trimethylstannylbenzoate (Me-STB), which was then reacted with murine 81C6 mAb without purification of the SAB intermediate. Radiochemical purity, immunoreactive fraction, sterility, and apyrogenicity of the 211At-labeled 81C6 preparations were evaluated. Results: Murine 81C6 mAb was successfully labeled with 211At using these revised procedures with improved radiochemical yields and decreased overall synthesis time compared with the original clinical labeling procedure. Conclusions: With 2.74 GBq of 211At, it was possible to produce 1.0 GBq of 211At-labeled 81C6 with an immunoreactive fraction of 92%. These revised procedures permit production of 211At-labeled mAbs suitable for use at clinically relevant activity levels.


Assuntos
Anticorpos Monoclonais/química , Astato/química , Neoplasias Encefálicas/terapia , Glioma/terapia , Marcação por Isótopo/métodos , Partículas alfa/uso terapêutico , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Astato/isolamento & purificação , Astato/farmacocinética , Astato/uso terapêutico , Benzoatos/química , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioma/imunologia , Glioma/patologia , Humanos , Camundongos , Radioimunoterapia/métodos , Distribuição Tecidual , Compostos de Trimetilestanho/química , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cancer Biother Radiopharm ; 35(6): 425-436, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32077749

RESUMO

Despite the consensus around the clinical potential of the α-emitting radionuclide astatine-211 (211At), there are only a limited number of research facilities that work with this nuclide. There are three main reasons for this: (1) Scarce availability of the nuclide. Despite a relatively large number of globally existing cyclotrons capable of producing 211At, few cyclotron facilities produce the nuclide on a regular basis. (2) Lack of a chemical infrastructure, that is, isolation of 211At from irradiated targets and the subsequent synthesis of an astatinated product. At present, the research groups that work with 211At depend on custom systems for recovering 211At from the irradiated targets. Setting up and implementing such custom units require long lead times to provide a proper working system. (3) The chemistry of 211At. Compared with radiometals there are no well-established and generally accepted synthesis methods for forming sufficiently stable bonds between 211At and the tumor-specific vector to allow for systemic applications. Herein we present an overview of the infrastructure of producing 211At radiopharmaceuticals, from target to radiolabeled product including chemical strategies to overcome hurdles for advancement into clinical trials with 211At.


Assuntos
Astato/química , Ciclotrons , Neoplasias/radioterapia , Radioterapia (Especialidade)/instrumentação , Compostos Radiofarmacêuticos/química , Partículas alfa/uso terapêutico , Astato/isolamento & purificação , Astato/uso terapêutico , Ensaios Clínicos como Assunto , Humanos , Radioterapia (Especialidade)/métodos , Compostos Radiofarmacêuticos/isolamento & purificação , Compostos Radiofarmacêuticos/uso terapêutico
15.
J Nucl Med ; 61(2): 242-248, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31451486

RESUMO

Targeted radionuclide therapy (TRT) targeting oncoproteins facilitates the delivery of therapeutic radionuclides to tumor tissues with high precision. Herein, we developed 2 new radiopharmaceuticals, 4-131I-iodo- and 4-211At-astato-N-[4-(6-(isopropylamino)pyridine-4-yl)-1,3-thiazol-2-yl]-N-methylbenzamide (131I-IITM and 211At-AITM), targeting the ectopic metabotropic glutamate receptor 1 (mGluR1) in melanomas for TRT studies. Methods:131I-IITM and 211At-AITM were synthesized by reacting a stannyl precursor with 131I-NaI and 211At in the presence of an oxidizing agent. The therapeutic efficacy and safety of the 2 radiopharmaceuticals were investigated using mGluR1-expressing B16F10 melanoma cells and melanoma-bearing mice. Results:131I-IITM and 211At-AITM were obtained with a radiochemical purity of greater than 99% and radiochemical yields of 42.7% ± 10.4% and 45.7% ± 6.5%, respectively, based on the total radioactivity of used radionuclides. 131I-IITM and 211At-AITM exhibited a maximum uptake of 4.66% ± 0.70 and 7.68% ± 0.71 percentage injected dose per gram (%ID/g) in the targeted melanomas, respectively, and were rapidly cleared from nontarget organs after intravenous injection. Both agents markedly inhibited melanoma growth compared with the controls (61.00% and 95.68%, respectively). In the melanoma model, considerably greater therapeutic efficacy with negligible toxicity was observed using 211At-AITM. Conclusion: The nontoxic radiopharmaceuticals 131I-IITM and 211At-AITM are useful high-precision TRT agents that can be used to target the oncoprotein mGluR1 for melanoma therapy.


Assuntos
Astato/uso terapêutico , Benzamidas/uso terapêutico , Radioisótopos do Iodo/uso terapêutico , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Benzamidas/efeitos adversos , Benzamidas/química , Benzamidas/farmacocinética , Melanoma Experimental/metabolismo , Melanoma Experimental/radioterapia , Camundongos , Radioquímica , Segurança , Distribuição Tecidual , Proteína Tumoral 1 Controlada por Tradução
16.
Blood ; 134(15): 1247-1256, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31395601

RESUMO

Minimal residual disease (MRD) has become an increasingly prevalent and important entity in multiple myeloma (MM). Despite deepening responses to frontline therapy, roughly 75% of MM patients never become MRD-negative to ≤10-5, which is concerning because MRD-negative status predicts significantly longer survival. MM is highly heterogeneous, and MRD persistence may reflect survival of isolated single cells and small clusters of treatment-resistant subclones. Virtually all MM clones are exquisitely sensitive to radiation, and the α-emitter astatine-211 (211At) deposits prodigious energy within 3 cell diameters, which is ideal for eliminating MRD if effectively targeted. CD38 is a proven MM target, and we conjugated 211At to an anti-CD38 monoclonal antibody to create an 211At-CD38 therapy. When examined in a bulky xenograft model of MM, single-dose 211At-CD38 at 15 to 45 µCi at least doubled median survival of mice relative to untreated controls (P < .003), but no mice achieved complete remission and all died within 75 days. In contrast, in a disseminated disease model designed to reflect low-burden MRD, 3 studies demonstrated that single-dose 211At-CD38 at 24 to 45 µCi produced sustained remission and long-term survival (>150 days) for 50% to 80% of mice, where all untreated mice died in 20 to 55 days (P < .0001). Treatment toxicities were transient and minimal. These data suggest that 211At-CD38 offers the potential to eliminate residual MM cell clones in low-disease-burden settings, including MRD. We are optimistic that, in a planned clinical trial, addition of 211At-CD38 to an autologous stem cell transplant (ASCT) conditioning regimen may improve ASCT outcomes for MM patients.


Assuntos
ADP-Ribosil Ciclase 1 , Astato/uso terapêutico , Imunoconjugados/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Neoplasia Residual/tratamento farmacológico , ADP-Ribosil Ciclase 1/análise , Astato/administração & dosagem , Astato/farmacocinética , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/farmacocinética , Masculino , Mieloma Múltiplo/patologia , Neoplasia Residual/patologia
18.
Appl Radiat Isot ; 139: 238-243, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29864741

RESUMO

Astatine-211 is a promising radionuclide for targeted radiotherapy. It is required to image the distribution of targeted radiotherapeutic agents in a patient's body for optimization of treatment strategies. We proposed to image 211At with high-energy photons to overcome some problems in conventional planar or single-photon emission computed tomography imaging. We performed an imaging experiment of a point-like 211At source using a Compton camera, and demonstrated the capability of imaging 211At with the high-energy photons for the first time.


Assuntos
Astato/uso terapêutico , Cintilografia/instrumentação , Radioterapia/instrumentação , Partículas alfa/uso terapêutico , Simulação por Computador , Humanos , Método de Monte Carlo , Neoplasias/diagnóstico por imagem , Neoplasias/radioterapia , Fótons , Cintilografia/estatística & dados numéricos , Compostos Radiofarmacêuticos/uso terapêutico , Radioterapia/estatística & dados numéricos
19.
Cancer Sci ; 109(7): 2302-2309, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29952132

RESUMO

Synovial sarcoma (SS) is a rare yet refractory soft-tissue sarcoma that predominantly affects young adults. We show in a mouse model that radioimmunotherapy (RIT) with an α-particle emitting anti-Frizzled homolog 10 (FZD10) antibody, synthesized using the α-emitter radionuclide astatine-211 (211 At-OTSA101), suppresses the growth of SS xenografts more efficiently than the corresponding ß-particle emitting anti-FZD10 antibody conjugated with the ß-emitter yettrium-90 (90 Y-OTSA101). In biodistribution analysis, 211 At was increased in the SS xenografts but decreased in other tissues up to 1 day after injection as time proceeded, albeit with a relatively higher uptake in the stomach. Single 211 At-OTSA101 doses of 25 and 50 µCi significantly suppressed SS tumor growth in vivo, whereas a 50-µCi dose of 90 Y-OTSA101 was needed to achieve this. Importantly, 50 µCi of 211 At-OTSA101 suppressed tumor growth immediately after injection, whereas this effect required several days in the case of 90 Y-OTSA101. Both radiolabeled antibodies at the 50-µCi dosage level significantly prolonged survival. Histopathologically, severe cellular damage accompanied by massive cell death was evident in the SS xenografts at even 1 day after the 211 At-OTSA101 injection, but these effects were relatively milder with 90 Y-OTSA101 at the same timepoint, even though the absorbed doses were comparable (3.3 and 3.0 Gy, respectively). We conclude that α-particle RIT with 211 At-OTSA101 is a potential new therapeutic option for SS.


Assuntos
Astato/uso terapêutico , Receptores Frizzled/antagonistas & inibidores , Radioimunoterapia/métodos , Radioisótopos/uso terapêutico , Sarcoma Sinovial/radioterapia , Partículas alfa/uso terapêutico , Animais , Anticorpos Monoclonais/uso terapêutico , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Molecules ; 23(3)2018 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-29510568

RESUMO

This review summarizes recent progress and developments as well as the most important pitfalls in targeted alpha-particle therapy, covering single alpha-particle emitters as well as in vivo alpha-particle generators. It discusses the production of radionuclides like 211At, 223Ra, 225Ac/213Bi, labelling and delivery employing various targeting vectors (small molecules, chelators for alpha-emitting nuclides and their biomolecular targets as well as nanocarriers), general radiopharmaceutical issues, preclinical studies, and clinical trials including the possibilities of therapy prognosis and follow-up imaging. Special attention is given to the nuclear recoil effect and its impacts on the possible use of alpha emitters for cancer treatment, proper dose estimation, and labelling chemistry. The most recent and important achievements in the development of alpha emitters carrying vectors for preclinical and clinical use are highlighted along with an outlook for future developments.


Assuntos
Actínio/uso terapêutico , Partículas alfa/uso terapêutico , Astato/uso terapêutico , Bismuto/uso terapêutico , Neoplasias/radioterapia , Radioisótopos/uso terapêutico , Compostos Radiofarmacêuticos/uso terapêutico , Rádio (Elemento)/uso terapêutico , Actínio/química , Astato/química , Bismuto/química , Quelantes/química , Quelantes/farmacocinética , Relação Dose-Resposta à Radiação , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Compostos Heterocíclicos/química , Compostos Heterocíclicos/farmacocinética , Compostos Heterocíclicos com 1 Anel/química , Compostos Heterocíclicos com 1 Anel/farmacocinética , Humanos , Neoplasias/patologia , Doses de Radiação , Radioisótopos/química , Compostos Radiofarmacêuticos/química , Rádio (Elemento)/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...