Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Mol Pharm ; 18(11): 4179-4187, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34591481

RESUMO

As an excellent target for cancer theranostics, fibroblast activation protein (FAP) has become an attractive focus in cancer research. A class of FAP inhibitors (FAPIs) with a N-(4-quinolinoyl)-Gly-(2-cyanopyrrolidine) scaffold were developed, which displayed nanomolar affinity and high selectivity. Compared with 90Y, 177Lu, 225Ac, and 188Re, 211At seems to be more favored as a therapeutic candidate for FAPI tracers which have fast washout and short retention in tumor sites. Thus, the current study reported the synthesis of two FAPI precursors for 211At and 131I labeling and the preliminary evaluation of 131I-labeled FAPI analogues for cancer theranostics. FAPI variants with stannyl precursors were successfully synthesized and labeled with 131I using a radioiododestannylation reaction. Two radioactive tracers were obtained with high radiochemical purity over 99% and good radiochemical yields of 58.2 ± 1.78 and 59.5 ± 4.44% for 131I-FAPI-02 and 131I-FAPI-04, respectively. Both tracers showed high specific binding to U87MG cells in comparison with little binding to MCF-7 cells. Compared to 131I-FAPI-02, 131I-FAPI-04 exhibited higher affinity, more intracellular uptake, and longer retention time in vitro. Biodistribution studies revealed that both tracers were mainly excreted through the kidneys as well as the hepatobiliary pathway due to their high lipophilicity. In addition, higher accumulation, longer dwell time, and increased tumor-to-organ ratios were achieved by 131I-FAPI-04, which was clearly demonstrated by SPECT/CT imaging. Furthermore, intratumor injection of 131I-FAPI-04 significantly suppressed the tumor growth in U87MG xenograft mice without significant toxicity observed. The above results implied that FAP-targeted alpha endoradiotherapy (specific to 211At) should be used to treat tumors in the near future, considering the chemical similarity between iodine and astatine can ensure the labeling of the latter onto the designed FAPIs.


Assuntos
Astato/administração & dosagem , Proteínas de Membrana/antagonistas & inibidores , Neoplasias/terapia , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Linhagem Celular Tumoral , Endopeptidases , Humanos , Radioisótopos do Iodo , Camundongos , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Traçadores Radioativos , Nanomedicina Teranóstica/métodos , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Cancer Sci ; 112(3): 1132-1140, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33277750

RESUMO

α-Methyl-l-tyrosine (AMT) has a high affinity for the cancer-specific l-type amino acid transporter 1 (LAT1). Therefore, we established an anti-cancer therapy, with 211 At-labeled α-methyl-l-tyrosine (211 At-AAMT) as a carrier of 211 At into tumors. 211 At-AAMT had high affinity for LAT1, inhibited tumor cell growth, and induced DNA double-stranded breaks in vitro. We evaluated the accumulation of 211 At-AAMT in vivo and the role of LAT1. Treatment with 0.4 MBq/mouse 211 At-AAMT inhibited tumor growth in the PANC-1 tumor model and 1 MBq/mouse 211 At-AAMT inhibited metastasis in the lung of the B16F10 metastasis model. Our results suggested that 211 At would be useful for anti-cancer therapy and that LAT1 is suitable as a target for radionuclide therapy.


Assuntos
Partículas alfa/uso terapêutico , Astato/administração & dosagem , Portadores de Fármacos/farmacologia , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Neoplasias/radioterapia , alfa-Metiltirosina/farmacologia , Animais , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Modelos Animais de Doenças , Estudos de Viabilidade , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
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
4.
Mol Pharm ; 16(8): 3524-3533, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31268724

RESUMO

The use of nanobodies (Nbs) as vehicles in targeted alpha therapy (TAT) has gained great interest because of their excellent properties. They combine high in vivo affinity and specificity of binding with fast kinetics. This research investigates a novel targeted therapy that combines the α-particle emitter astatine-211 (211At) and the anti-HER2 Nb 2Rs15d to selectively target HER2+ cancer cells. Two distinctive radiochemical methodologies are investigated using three different coupling reagents. The first method uses the coupling reagents, N-succinimidyl 4-(1,2-bis-tert-butoxycarbonyl)guanidinomethyl-3-(trimethylstannyl)benzoate (Boc2-SGMTB) and N-succinimidyl-3-(trimethylstannyl)benzoate (m-MeATE), which are both directed to amino groups on the Nb, resulting in random conjugation. The second method aims at obtaining a homogeneous tracer population, via a site-specific conjugation of the N-[2-(maleimido)ethyl]-3-(trimethylstannyl)benzamide (MSB) reagent onto the carboxyl-terminal cysteine of the Nb. The resulting radioconjugates are evaluated in vitro and in vivo. 2Rs15d is labeled with 211At using Boc2-SGMTB, m-MeATE, and MSB. After astatination and purification, the binding specificity of the radioconjugates is validated on HER2+ cells, followed by an in vivo biodistribution assessment in SKOV-3 xenografted mice. α-camera imaging is performed to determine uptake and activity distribution in kidneys/tumors. 2Rs15d astatination resulted in a high radiochemical purity >95% for all radioconjugates. The biodistribution studies of all radioconjugates revealed comparable tumor uptake (higher than 8% ID/g at 1 h). [211At]SAGMB-2Rs15d showed minor uptake in normal tissues. Only in the kidneys, a higher uptake was measured after 1 h, but decreased rapidly after 3 h. Astatinated Nbs consisting of m-MeATE or MSB reagents revealed elevated uptake in lungs and stomach, indicating the presence of released 211At. α-Camera imaging of tumors revealed a homogeneous activity distribution. The radioactivity in the kidneys was initially concentrated in the renal cortex, while after 3 h most radioactivity was measured in the medulla, confirming the fast washout into urine. Changing the reagents for Nb astatination resulted in different in vivo biodistribution profiles, while keeping the targeting moiety identical. Boc2-SGMTB is the preferred reagent for Nb astatination because of its high tumor uptake, its low background signals, and its fast renal excretion. We envision [211At]SAGMB-2Rs15d to be a promising therapeutic agent for TAT and aim toward efficacy evaluation.


Assuntos
Astato/administração & dosagem , Imunoconjugados/administração & dosagem , Neoplasias Ovarianas/radioterapia , Receptor ErbB-2/antagonistas & inibidores , Anticorpos de Domínio Único/administração & dosagem , Partículas alfa/uso terapêutico , Animais , Astato/química , Astato/farmacocinética , Benzoatos/química , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Feminino , Humanos , Imunoconjugados/química , Imunoconjugados/imunologia , Imunoconjugados/farmacocinética , Camundongos , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/patologia , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/imunologia , Distribuição Tecidual , Compostos de Trimetilestanho/química , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Nucl Med ; 58(2): 346-353, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27765860

RESUMO

Targeted α-therapy is a promising treatment option for various types of malignant tumors. Radiolabeled cancer-seeking agents, however, undergo degradation, resulting in a certain percentage of free radionuclide in the body. The radiohalogen 211At accumulates in various tissues, with specifically high uptake in the thyroid. When normal thyroid function is disturbed because of ionizing radiation (IR) exposure, deleterious effects can occur in tissues that depend on thyroid hormone (TH) regulation for normal physiologic function. However, knowledge of systemic effects is still rudimentary. We previously reported similarities in transcriptomic regulation between the thyroid and other tissues despite large differences in absorbed dose from 211At. Here, we present supportive evidence on systemic effects after 211At administration. METHODS: Expression microarray data from the kidney cortex and medulla, liver, lungs, and spleen were used from previous studies in which mice were intravenously injected with 0.064-42 kBq of 211At and killed after 24 h or injected with 1.7 kBq of 211At and killed after 1, 6, or 168 h. Controls were mock-treated and killed after 24 h. Literature-based gene signatures were used to evaluate the relative impact from IR- or TH-induced regulation. Thyroid- and TH-associated upstream regulators as well as thyroid-related diseases and functions were generated using functional analysis software. RESULTS: Responses in IR- or TH-associated gene signatures were tissue-specific and varied over time, and the relative impact of each gene signature differed between the investigated tissues. The liver showed a clear dominance of TH-responding genes. In the kidney cortex, kidney medulla, and lungs, the TH-associated signature was detected to at least an extent similar to the IR-associated signature. The spleen was the single tissue showing regulation of only IR-associated signature genes. Various thyroid-associated diseases and functions were inferred from the data: L-triiodothyronine, TH, TH receptor, and triiodothyronine (reverse) were inferred as upstream regulators with differences in incidence and strength of regulation depending on tissue type. CONCLUSION: These findings indicate that transcriptional regulation in various nonthyroid tissues was-in part-induced by thyroid (hormone)-dependent signaling. Consideration of the systemic context between tissues could contribute to normal tissue risk assessment and planning of remedial measures.


Assuntos
Astato/administração & dosagem , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/efeitos da radiação , Especificidade de Órgãos/genética , Ativação Transcricional/genética , Ativação Transcricional/efeitos da radiação , Animais , Relação Dose-Resposta à Radiação , Camundongos Endogâmicos BALB C , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Especificidade de Órgãos/efeitos da radiação , Doses de Radiação , Compostos Radiofarmacêuticos/efeitos da radiação , Glândula Tireoide/fisiologia , Glândula Tireoide/efeitos da radiação
6.
Cancer Biother Radiopharm ; 29(6): 238-46, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24971673

RESUMO

UNLABELLED: Alpha-particle emitters, such as astatine-211 (211At), are generally considered suitable for the treatment of small cell clusters due to their short path length, while beta-particle emitters, for example, Lutetium-177 (177Lu), have a longer path length and are considered better for small, established tumors. A combination of such radionuclides may be successful in regimens of radioimmunotherapy. In this study, rats were treated by sequential administration of first a 177Lu-labeled antibody, followed by a 211At-labeled antibody 25 days later. METHODS: Rats bearing solid colon carcinoma tumors were treated with 400 MBq/kg body weight 177Lu-BR96. After 25 days, three groups of animals were given either 5 or 10 MBq/kg body weight of 211At-BR96 simultaneously with or without a blocking agent reducing halogen uptake in normal tissues. Control animals were not given any 211At-BR96. Myelotoxicity, body weight, tumor size, and development of metastases were monitored for 120 days. RESULTS: Tumors were undetectable in 90% of the animals on day 25, independent of treatment. Additional treatment with 211At-labeled antibodies did not reduce the proportion of animals developing metastases. The rats suffered from reversible myelotoxicity after treatment. CONCLUSIONS: Sequential administration of 177Lu-BR96 and 211At-BR96 resulted in tolerable toxicity providing halogen blocking but did not enhance the therapeutic effect.


Assuntos
Astato/administração & dosagem , Neoplasias do Colo/radioterapia , Lutécio/administração & dosagem , Radioimunoterapia/métodos , Radioisótopos/administração & dosagem , Partículas alfa , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Humanos , Compostos Radiofarmacêuticos/administração & dosagem , Ratos
7.
Nuklearmedizin ; 52(6): 212-21, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24036694

RESUMO

OBJECTIVE: Increased amino acid transport in brain tumours is used for diagnostic purposes. It has been shown that the α-emitting radionuclide astatine-211 labeled to L-phenylalanine is taken up by glioblastoma cells. We here tested, if systemic treatment with 4-[211At]astatine-phenylalanine (At-Phe) has a beneficial effect on survival of rats with intracranial glioblastoma. ANIMALS, METHODS: The rat glioblastoma cell line BT4Ca was implanted into the prefrontal cortex of female BDIX rats by stereotaxic microinjection (10,000 cells/3 µl; n = 83). 3 days after implantation At-Phe or phosphate buffered saline were injected intravenously. A third group was treated twice, i.e., on day 3 and 10. Health condition was assessed each day by using a score system. Rats were sacrificed on days 6, 10, 13 and 17 after implantation, or when showing premortal health condition to measure tumour volume and necrosis. The proliferation index (PI) was assessed after immunohistochemical staining of Ki-67. RESULTS: Survival time of rats treated twice with At-Phe was significantly prolonged. Additionally, both At-Phe-treated groups remained significantly longer in a better health condition. Rats with poor health status had larger tumours than rats with fair health condition. Overall, irrespective of treatment the PI was reduced in rats with poor health condition. Necrosis was larger in rats treated twice with At-Phe. CONCLUSION: Intravenous treatment with At-Phe enhanced survival time of rats with intracranial glioblastomas and improved health condition. These results encourage studies using local treatment of intracranial glioblastoma with At-Phe, either by repeated local injection or by intracavital application after tumour resection.


Assuntos
Astato/administração & dosagem , Braquiterapia/métodos , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Fenilalanina/administração & dosagem , Animais , Neoplasias Encefálicas/diagnóstico , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Feminino , Glioblastoma/diagnóstico , Compostos Radiofarmacêuticos/administração & dosagem , Ratos , Taxa de Sobrevida , Resultado do Tratamento
8.
J Nucl Med ; 54(6): 990-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23658216

RESUMO

UNLABELLED: (211)At is a promising therapeutic radionuclide because of the nearly optimal biological effectiveness of emitted α-particles. Unbound (211)At accumulates in the thyroid gland and in other vital normal tissues. However, few studies have been performed that assess the (211)At-induced normal-tissue damage in vivo. Knowledge about the extent and quality of resulting responses in various organs offers a new venue for reducing risks and side effects and increasing the overall well-being of the patient during and after therapy. METHODS: Female BALB/c nude mice were injected intravenously with 0.064-42 kBq of (211)At or mock-treated, and the kidneys, liver, lungs, and spleen were excised 24 h after injection. A transcriptional gene expression analysis was performed in triplicate using RNA microarray technology. Biological processes associated with regulated transcripts were grouped into 8 main categories with 31 subcategories according to gene ontology terms for comparison of regulatory profiles. RESULTS: A substantial decrease in the total number of regulated transcripts was observed between 0.64 and 1.8 kBq of (211)At for all investigated tissues. Few genes were differentially regulated in each tissue at all absorbed doses. In all tissues, most of these genes showed a nonmonotonous dependence on absorbed dose. However, the direction of regulation generally remained uniform for a given gene. Few known radiation-associated genes were regulated on the transcriptional level, and their expression profile generally appeared to be dose-independent and tissue-specific. The regulatory profiles of categorized biological processes were tissue-specific and reflected the shift in regulatory intensity between 0.64 and 1.8 kBq of (211)At. The profiles revealed strongly regulated and nonregulated subcategories. CONCLUSION: The strong regulatory change observed between 0.64 and 1.8 kBq is hypothesized to result not only from low-dose effects in each tissue but also from physiologic responses to ionizing radiation-induced damage to, for example, the (211)At-accumulating thyroid gland. The presented results demonstrate the complexity of responses to radionuclides in vivo and highlight the need for further research to also consider physiology in ionizing radiation-induced responses.


Assuntos
Astato/administração & dosagem , Astato/efeitos adversos , Regulação da Expressão Gênica/efeitos da radiação , Transcrição Gênica/efeitos da radiação , Administração Intravenosa , Partículas alfa/efeitos adversos , Partículas alfa/uso terapêutico , Animais , Astato/uso terapêutico , Relação Dose-Resposta à Radiação , Feminino , Camundongos , Especificidade de Órgãos , Proteção Radiológica
9.
Cancer Biother Radiopharm ; 28(2): 108-14, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23230896

RESUMO

UNLABELLED: Abstract Purpose: Pretargeted radioimmunotherapy (PRIT) against intraperitoneal (i.p.) ovarian microtumors using avidin-conjugated monoclonal antibody MX35 (avidin-MX35) and (211)At-labeled, biotinylated, succinylated poly-l-lysine ((211)At-B-PLsuc) was compared with conventional radioimmunotherapy (RIT) using (211)At-labeled MX35 in a nude mouse model. METHODS: Mice were inoculated i.p. with 1×10(7) NIH:OVCAR-3 cells. After 3 weeks, they received PRIT (1.0 or 1.5 MBq), RIT (0.9 MBq), or no treatment. Concurrently, 10 additional animals were sacrificed and examined to determine disease progression at the start of therapy. Treated animals were analyzed with regard to presence of tumors and ascites (tumor-free fraction; TFF), 8 weeks after therapy. RESULTS: Tumor status at baseline was advanced: 70% of sacrificed animals exhibited ascites. The TFFs were 0.35 (PRIT 1.0 MBq), 0.45 (PRIT 1.5 MBq), and 0.45 (RIT). The 1.5-MBq PRIT group exhibited lower incidence of ascites and fewer tumors >1 mm than RIT-treated animals. CONCLUSIONS: PRIT was as effective as RIT with regard to TFF; however, the size distribution of tumors and presence of ascites indicated that 1.5-MBq PRIT was more efficient. Despite advanced disease in many animals at the time of treatment, PRIT demonstrated good potential to treat disseminated ovarian cancer.


Assuntos
Partículas alfa/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Astato/administração & dosagem , Avidina/uso terapêutico , Modelos Animais de Doenças , Neoplasias Ovarianas/radioterapia , Radioimunoterapia , Animais , Anticorpos Monoclonais/farmacocinética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/imunologia , Cintilografia , Distribuição Tecidual
10.
Cancer Biother Radiopharm ; 26(6): 727-36, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22087606

RESUMO

PURPOSE: Avidin-coupled monoclonal antibody MX35 (avidin-MX35) and astatine-211-labeled, biotinylated, succinylated poly-l-lysine ((211)At-B-PL(suc)) were administered in mice to assess potential efficacy as an intraperitoneal (i.p.) therapy for microscopic tumors. We aimed to establish a timeline for pretargeted radioimmunotherapy using these substances, and estimate the maximum tolerable activity. METHODS: (125)I-avidin-MX35 and (211)At-B-PL(suc) were administered i.p. in nude mice. Tissue distributions were studied at various time points and mean absorbed doses were estimated from organ uptake of (211)At-B-PL(suc). Studies of myelotoxicity were performed after administration of different activities of (211)At-B-PL(suc). RESULTS: We observed low blood content of both (125)I-avidin-MX35 and (211)At-B-PL(suc), indicating fast clearance. After sodium perchlorate blocking, the highest (211)At uptake was found in kidneys. Red bone marrow (RBM) accumulated some (211)At activity. Mean absorbed doses of special interest were 2.3 Gy/MBq for kidneys, 0.4 Gy/MBq for blood, and 0.9 Gy/MBq for RBM. An absorbed dose of 0.9 Gy to the RBM was found to be safe. These values suggested that RBM would be the key dose-limiting organ in the proposed pretargeting scheme, and that blood data alone was not sufficient for predicting its absorbed dose. CONCLUSIONS: To attain a favorable distribution of activity and avoid major toxicities, at least 1.0 MBq of (211)At-B-PL(suc) can be administered 24 hours after an i.p. injection of avidin-MX35. These results provide a basis for future i.p. therapy studies in mice of microscopic ovarian cancer.


Assuntos
Anticorpos Monoclonais/farmacocinética , Astato/farmacocinética , Avidina/farmacocinética , Lisina/farmacocinética , Radioimunoterapia/métodos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Astato/administração & dosagem , Astato/química , Avidina/administração & dosagem , Avidina/química , Biotinilação/métodos , Medula Óssea/efeitos dos fármacos , Feminino , Isótopos de Iodo/administração & dosagem , Isótopos de Iodo/química , Isótopos de Iodo/farmacocinética , Marcação por Isótopo/métodos , Rim/efeitos dos fármacos , Lisina/administração & dosagem , Lisina/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/radioterapia , Polímeros/administração & dosagem , Polímeros/química , Polímeros/farmacocinética , Distribuição Tecidual
11.
J Nucl Med ; 50(7): 1161-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19525464

RESUMO

UNLABELLED: Recent clinical and experimental data demonstrate that thyroid stunning is caused by previous irradiation and may influence the efficacy of (131)I radiation therapy of thyroid cancer and possibly hyperthyroidism. To avoid stunning, many clinics have exchanged (131)I for (123)I for pretherapeutic diagnostic imaging and dose planning. Furthermore, recent in vitro studies indicate that (131)I irradiation reduces iodide uptake by downregulating the expression of the sodium iodide symporter (NIS). The rationale for this study was therefore to study effects on iodide transport and NIS messenger RNA (mRNA) expression in thyrocytes exposed to both (123)I and (131)I in addition to some other potentially interesting radionuclides. METHODS: Thyrotropin-stimulated thyroid cell monolayers were exposed to 0.5 Gy of (123)I, (131)I, (99m)Tc, or (211)At, all being radionuclides transported via NIS, in the culture medium for 6 h, or to various absorbed doses of (123)I or (131)I for 48 h. NIS mRNA expression was analyzed using quantitative reverse-transcriptase polymerase chain reaction. RESULTS: Iodide transport and NIS mRNA expression were reduced by all radionuclides. At the same absorbed dose, iodide transport was reduced the most by (211)At, followed by (123)I and (99m)Tc (equally potent), whereas (131)I was least effective. The onset of NIS downregulation was rapid (<1 d after irradiation) in cells exposed to (123)I or (211)At and was delayed in cells irradiated with (131)I or (99m)Tc. Iodide transport and NIS expression were recovered only for (211)At. (123)I reduced the iodine transport and the NIS mRNA expression more efficiently than did (131)I at an equivalent absorbed dose, with a relative biological effectiveness of about 5. CONCLUSION: The stunning effect per unit absorbed dose is more severe for (123)I than for (131)I. Despite the lower absorbed dose per unit activity for (123)I than for (131)I, stunning by (123)I cannot be excluded in patients. The degree to which iodide transport capacity and NIS mRNA expression are reduced seems to be related to the biological effectiveness of the type of radiation delivering the absorbed dose to the target, with (211)At (which has the highest relative biological effectiveness) causing the highest degree of stunning per unit absorbed dose in the present study.


Assuntos
Astato/administração & dosagem , Radioisótopos do Iodo/administração & dosagem , Iodo/metabolismo , Simportadores/metabolismo , Tecnécio/administração & dosagem , Doenças da Glândula Tireoide/metabolismo , Glândula Tireoide/metabolismo , Animais , Transporte Biológico Ativo/efeitos da radiação , Células Cultivadas , Relação Dose-Resposta à Radiação , Regulação da Expressão Gênica/efeitos da radiação , RNA Mensageiro/genética , Doses de Radiação , Compostos Radiofarmacêuticos/administração & dosagem , Suínos , Simportadores/genética , Glândula Tireoide/efeitos dos fármacos
12.
Eur J Nucl Med Mol Imaging ; 35(7): 1272-81, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18404268

RESUMO

PURPOSE: We reported recently the induction of selective iodide uptake in prostate cancer cells (LNCaP) by prostate-specific antigen (PSA) promoter-directed sodium iodide symporter (NIS) expression that allowed a significant therapeutic effect of (131)I. In the current study, we studied the potential of the high-energy alpha-emitter (211)At, also transported by NIS, as an alternative radionuclide after NIS gene transfer in tumors with limited therapeutic efficacy of (131)I due to rapid iodide efflux. METHODS: We investigated uptake and therapeutic efficacy of (211)At in LNCaP cells stably expressing NIS under the control of the PSA promoter (NP-1) in vitro and in vivo. RESULTS: NP-1 cells concentrated (211)At in a perchlorate-sensitive manner, which allowed a dramatic therapeutic effect in vitro. After intraperitoneal injection of (211)At (1 MBq), NP-1 tumors accumulated approximately 16% ID/g (211)At (effective half-life 4.6 h), which resulted in a tumor-absorbed dose of 1,580+/-345 mGy/MBq and a significant tumor volume reduction of up to 82+/-19%, while control tumors continued their growth exponentially. CONCLUSIONS: A significant therapeutic effect of (211)At has been demonstrated in prostate cancer after PSA promoter-directed NIS gene transfer in vitro and in vivo suggesting a potential role for (211)At as an attractive alternative radioisotope for NIS-targeted radionuclide therapy, in particular in smaller tumors with limited radionuclide retention time.


Assuntos
Astato/uso terapêutico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/radioterapia , Simportadores/metabolismo , Animais , Astato/administração & dosagem , Astato/farmacocinética , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Regiões Promotoras Genéticas , Antígeno Prostático Específico/genética , Neoplasias da Próstata/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Simportadores/genética , Transfecção , Transplante Heterólogo , Ensaio Tumoral de Célula-Tronco
13.
Int J Radiat Oncol Biol Phys ; 69(2): 572-9, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17869670

RESUMO

PURPOSE: To investigate the potential use of astatine-211 (211At)-labeled trastuzumab for the treatment of HER-2-positive, radioresistant ovarian carcinoma. METHODS AND MATERIALS: Four-week-old nude mice were inoculated intraperitoneally with 5 . 10(6) SKOV-3 cells in 0.4 mL saline on Day 0. The endpoint was the total tumor weight in each mouse on Day 63. Three experiments were performed in which the response to single-dose and fractionated treatment with unlabeled and 211At-labeled antibody was evaluated. RESULTS: Experiment 1 showed, for the same total amount of trastuzumab, a dose-response relationship between 211At activity (0-400 kBq on Day 7) and therapeutic efficacy (p = 0.001). The effect of varying the amount of unlabeled trastuzumab was studied in Experiment 2. All mice, except for the controls, received 400 kBq 211At-trastuzumab, and different groups received 5, 50, or 500 microg trastuzumab on Day 7. The increase from 5 to 50 microg trastuzumab reduced the tumors by 78% in weight. No tumors were present in mice given 500 microg trastuzumab. In Experiment 3, the effect of a fractionated treatment regimen was studied. Mice that received 100 kBq 211At-trastuzumab on Days 7 and 8 had a 42% smaller tumor burden than did controls. Groups of mice injected with 200 + 100 kBq on Days 7 and 21 and mice injected with 100 kBq on Days 7, 8, and 21 both had 24% less tumor weight than the corresponding controls. CONCLUSION: The combination of 500 microg trastuzumab and 400 kBq 211At-trastuzumab had the greatest effect, with complete eradication of the tumors in this nude mouse model.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Astato/uso terapêutico , Neoplasias Ovarianas/radioterapia , Tolerância a Radiação , Radioimunoterapia/métodos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Antineoplásicos/administração & dosagem , Astato/administração & dosagem , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/química , Neoplasias Ovarianas/patologia , Dosagem Radioterapêutica , Receptor ErbB-2 , Trastuzumab
14.
Eur J Nucl Med Mol Imaging ; 32(11): 1296-304, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16028065

RESUMO

PURPOSE: The purpose of this study was to analyse the properties of the astatinated chimeric MAb (cMAb) U36 as a conjugate to selectively target and eradicate head and neck squamous cell carcinoma (HNSCC). METHODS: cMAb U36 was labelled with 211At via the linker N-succinimidyl 4-(trimethylstannyl)benzoate (SPMB). The quality of the conjugate was extensively evaluated for binding and internalisation capacity, and compared with 125I-SPMB-cMAb U36. The cellular toxicity of the astatinated conjugate was assessed in two types of in vitro growth assay and compared with 131I-labelled cMAb U36 (directly labelled). RESULTS: Comparisons between 211At-cMAb U36 and 125I-cMAb U36 demonstrated an optimal functional capacity of the labelled products. Immunoreactivity and affinity assays showed high immunoreactive fractions (>93%), and an affinity in good agreement between the astatinated and iodinated antibodies. For both conjugates, specific binding to HNSCC cells could be demonstrated, as well as some internalisation. Retention of the astatinated conjugate was just slightly lower than for the iodinated conjugate and still reasonable for therapeutic use (31+/-2% vs 42.6+/-1.0% at 22 h), demonstrating no adverse effects from astatination of the antibody. Studies on cellular toxicity demonstrated a dose-dependent and antigen-specific cellular toxicity for 211At-cMAb U36, with about 10% cell survival at 50 decays per cell. The 131I-labelled conjugate was not as efficient, with a surviving cell fraction of about 50% at 55 decays per cell. CONCLUSION: These results indicate that 211At-cMAb U36 might be a promising future candidate for eradicating HNSCC micrometastases in vivo.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Sobrevivência Celular/efeitos da radiação , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Anticorpos Monoclonais/uso terapêutico , Astato/administração & dosagem , Astato/farmacocinética , Astato/uso terapêutico , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Estudos de Viabilidade , Humanos , Radioimunoterapia/métodos
15.
Nuklearmedizin ; 43(2): 63-8, 2004 Mar.
Artigo em Alemão | MEDLINE | ID: mdl-15029267

RESUMO

AIM: Determination of the biological effect of the alpha emitter (211)At on cellular level as well as the assessment of dosimetric data in a tumour model in vivo. METHODS: Transplantation of malignant ascitic cells in mice intraperitoneally and estimation of tumour characteristics (doubling time of the cells, mean survival of the animals following an i.p. application of a defined tumour cell number). (211)At labelled human serum albumin microspheres B-20 (MSP) of varying activity were injected into tumour bearing mice intraperitoneally. The effectiveness of the therapy was evaluated by means of determination of the duration of cell cycle arrest as well as the microscopic analysis of the rate of abnormal mitotic cells due to radiation induced damage. Furthermore, dose dependence of survival was evaluated. RESULTS: Three days following the intraperitoneally application of 8 x 10(6) tumour cells, 50-600 kBq (211)At-MSP were applied into the abdominal cavity. Considering the volume of ascites at this time and the administered activity, dose calculations were performed. An activity of 50 kBq caused a dose of 0.84 Gy. The increase of radiation induced effect on ascitic tumour cells was correlated with the dose. Between the duration of the cell cycle arrest and the administered activity, a directly proportional correlation was found. The mean survival of non-treated animals was 16.9 +/- 3.7 days. The prolongation of the survival was proportional to the activity administered. Using a dosage of 10 Gy, five animals out of 16 survived. CONCLUSION: Therapy of malignant ascitic cells using (211)At-MSP was effective in vivo. For tumour therapy, the (211)At represents a highly effective alternative to usually applied beta emitters.


Assuntos
Astato/uso terapêutico , Carcinoma de Ehrlich/radioterapia , Animais , Astato/administração & dosagem , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Portadores de Fármacos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos CBA , Microesferas , Albumina Sérica
16.
Radiat Res ; 157(6): 633-41, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12005541

RESUMO

Astatine-211, an alpha-particle emitter, was employed in a model system for vascular-targeted radioimmunotherapy of small tumors in mouse lung to compare its performance relative to other radioisotopes in the same system. Astatine-211 was coupled to the lung blood vessel-targeting monoclonal antibody 201B with N-succinimidyl N-(4-[211At]astatophenethyl) succinamate linker. Biodistribution data showed that the conjugate delivered 211At to the lung (260-418% ID/g), where it remained with a biological half-time of about 30 h. BALB/c mice bearing about 100 lung tumor colonies of EMT-6 cells, each about 2000 cells in size, were treated with 211At-labeled monoclonal antibody 201B. The administered activity of 185 kBq per animal extended the life span of treated mice over untreated controls. Injections of 370 kBq, corresponding to an absorbed dose of 25-40 Gy, were necessary to eradicate all of the lung tumors. Mice receiving 740 kBq of 211At-labeled monoclonal antibody 201B developed pulmonary fibrosis 3-4 months after treatment, as did mice treated with 3700 kBq of the alpha-particle emitter 213Bi-labeled monoclonal antibody 201B in previous work. Animals that were injected with 211At bound to untargeted IgG or to glycine, as control agents, also demonstrated therapeutic effects relative to untreated controls. Control groups that received untargeted 211At required about twice as much administered activity for effective therapy as did groups with lung-targeted radioisotope. These results were not consistent with radioisotope biodistribution and dosimetry calculations that indicated that lung-targeted 211At should be at least 10-fold more efficient for lung colony therapy than 211At bound to nontargeting controls. The data showed that 211At is useful for vascular-targeted radioimmunotherapy because lung tumor colonies were eradicated in the mice. Work in this model system demonstrates that vascular targeting of alpha-particle emitters is an efficient therapy for small perivascular tumors and may be applicable to human disease when specific targeting agents are identified.


Assuntos
Partículas alfa/uso terapêutico , Astato/uso terapêutico , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/radioterapia , Radioimunoterapia/métodos , Animais , Astato/administração & dosagem , Astato/metabolismo , Astato/farmacocinética , Relação Dose-Resposta à Radiação , Fibrose/radioterapia , Camundongos , Camundongos Endogâmicos BALB C , Especificidade de Órgãos , Tolerância a Radiação , Radioisótopos/administração & dosagem , Radioisótopos/metabolismo , Radioisótopos/farmacocinética , Radioisótopos/uso terapêutico , Solubilidade , Taxa de Sobrevida , Fatores de Tempo
17.
Acta Oncol ; 39(6): 741-5, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11130014

RESUMO

The purpose of the present study was to investigate the therapeutic efficacy of 211At-labelled specific monoclonal antibody MOv18 in nude mice with intraperitoneal growth of the human ovarian cancer cell line OVCAR3. In the first part of the study the antibody was injected intraperitoneally when the cancer growth was microscopic. The injected activity was 485-555 kBq. The median survival for treated mice was 213 days compared to 138 days for untreated mice (p < 0.014, log-rank test). No obvious toxicity was seen. Thirty-three percent of the mice were apparently free of cancer after 7 months and were probably cured. In the second part of the study mice with macroscopic cancer and signs of ascites were injected intraperitoneally with the same 211At-labelled antibody (377-389 kBq). This treatment possibly delayed the production of ascites. Hopefully radioimmunotherapy with regionally administered 211At-labelled antibody will be of value in women with ovarian cancer as well.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Astato/uso terapêutico , Imunoconjugados/farmacologia , Neoplasias Ovarianas/radioterapia , Neoplasias Peritoneais/radioterapia , Radioisótopos/uso terapêutico , Animais , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/imunologia , Ascite , Astato/administração & dosagem , Astato/farmacocinética , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/veterinária , Cuidados Paliativos , Neoplasias Peritoneais/imunologia , Neoplasias Peritoneais/veterinária , Radioisótopos/administração & dosagem , Radioisótopos/farmacocinética , Transplante Heterólogo , Células Tumorais Cultivadas
18.
Anticancer Res ; 20(1A): 459-62, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10769696

RESUMO

The aim of this study was to investigate the therapeutic efficacy of 211At-labelled monoclonal antibody given intraperitoneally to nude mice with intraperitoneal growth of a human ovarian cancer cell line. Female nude mice were inoculated intraperitoneally with 1 x 10(7) cells of the human ovarian cancer cell line NIH:OVCAR 3. After about two weeks they were injected with the 211At-labelled specific monoclonal antibody MOv18 intraperitoneally. For comparison, other groups of mice were given the same labelled antibody intravenously, 211At-labelled unspecific antibody C242 intraperitoneally or unalbelled MOv18 intraperitoneally. Six weeks later the animals were sacrificed and the occurrence of tumour and ascites was determined. When the mice were treated with 211At-labelled MOv18 intraperitoneally 9 out of 10 were apparently free of both ascites and tumour compared to none of the mice given unlabelled antibody. 211At-labelled MOv18 given intravenously or 211At-labelled unspecific antibody given intraperitoneally were less effective. Regional radioimmunotherapy with the alpa-emitter 211Astatine seems to be an effective treatment of nude mice with intraperitoneally growing human ovarian cancer. Hopefully this treatment can be given in an adjuvant setting to women with minimal residual ovarian cancer in the future.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Astato/uso terapêutico , Carcinoma/radioterapia , Imunoconjugados/uso terapêutico , Neoplasias Ovarianas/radioterapia , Neoplasias Peritoneais/radioterapia , Radioimunoterapia , Receptores de Superfície Celular , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Antineoplásicos/administração & dosagem , Antígenos de Neoplasias/imunologia , Ascite/etiologia , Ascite/radioterapia , Astato/administração & dosagem , Carcinoma/imunologia , Carcinoma/patologia , Proteínas de Transporte/imunologia , Feminino , Receptores de Folato com Âncoras de GPI , Humanos , Injeções Intraperitoneais , Injeções Intravenosas , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/imunologia , Transplante de Neoplasias , Neoplasia Residual , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/imunologia , Neoplasias Peritoneais/patologia , Transplante Heterólogo
19.
Int J Radiat Oncol Biol Phys ; 45(2): 491-9, 1999 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10487576

RESUMO

PURPOSE: The antitenascin human/mouse chimeric monoclonal antibody labeled with the alpha-particle-emitting radionuclide 211At is of interest as an endoradiotherapeutic agent for the treatment of brain tumors. To facilitate the investigation of 211At-labeled chimeric 81C6 in patients, the long-term radiotoxicity of this radiopharmaceutical has been evaluated. METHODS AND MATERIALS: Antibody labeling was performed using N-succinimidyl 3-[211At]astato-benzoate. After an initial dose-finding experiment, a second toxicity study was carried out at 4 dose levels in groups of 30 nonthyroid blocked B6C3F1 mice per group (15 males, 15 females). Male mice received either saline or 15-81 kBq/g and females received either saline or 16-83 kBq/g of 211At-labeled antibody. Ten animals (5 males, 5 females) were followed for 6 months and the remainder for 1 year. RESULTS: The lethal dose in 10% of animals (LD10) for 211At-labeled chimeric 81C6 was 46 kBq/g in females and 102 kBq/g in males. Toxic effects--perivascular fibrosis of the intraventricular septum of the heart, bone marrow suppression, splenic white pulp atrophy, and spermatic maturational delay--generally were confined to a few animals receiving the highest doses of labeled antibody. CONCLUSIONS: The LD10 of 211At-labeled chimeric 81C6 in this mouse strain was about half that of [211At]astatide. These results establish the preclinical maximum tolerated dose of 211At-labeled chimeric 81C6 and define in the mouse the target organs for toxicity. These studies will be useful for determining starting doses for clinical studies with 211At-labeled chimeric 81C6.


Assuntos
Astato/uso terapêutico , Imunoglobulina G/uso terapêutico , Imunotoxinas/uso terapêutico , Compostos Radiofarmacêuticos/uso terapêutico , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Astato/administração & dosagem , Astato/imunologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/imunologia , Imunotoxinas/administração & dosagem , Masculino , Camundongos , Especificidade de Órgãos , Compostos Radiofarmacêuticos/administração & dosagem , Fatores Sexuais
20.
Br J Cancer ; 77(7): 1115-22, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9569048

RESUMO

To determine the effects of 211At-labelled antibodies in solid tumour tissue, nude mice carrying OHS human osteosarcoma xenografts received intratumour injections at dosages of 1, 2 or 4 MBq (-1) tumour. The radioisotope was conjugated to either the osteosarcoma-specific monoclonal antibody TP-3 or the non-specific polyclonal antibody hlgGkappa. Tumour retention of injected radioimmunoconjugate (RIC), measured as the percentage of injected activity dosage per gram, was significantly higher for the [211At]TP-3 (203 +/- 93 at 24.1 h post injection) compared with the [211At]hlgGkappa (57 +/- 22 at 23.2 h post injection). The radioactive count rates in body (measured at neck and abdomen) were significantly lower with the TP-3 than with the hlgGkappa. Microautoradiography of the tumour radionuclide distribution was different for the two RICs, i.e. the [211At]TP-3 was to a larger extent concentrated near the injection site, whereas the [211At]hlgGkappa was more evenly distributed all over the tumour. The tumour growth was significantly delayed as a function of the injected activity dosage but without significant difference between the specific and the non-specific RIC. According to this study, it is possible to deliver highly selective radiation doses to solid tumours using intratumour injection of alpha-particle-emitting RICs. Improved tumour retention caused by antigen binding indicates that reduced normal tissue exposure can be obtained with antigen-specific antibodies. The heterogeneous tumour dose distribution observed is, however, a major impediment to the use of alpha-particle emitters against solid tumours.


Assuntos
Astato/administração & dosagem , Imunoglobulinas/administração & dosagem , Osteossarcoma/radioterapia , Radioimunoterapia/métodos , Animais , Astato/farmacocinética , Autorradiografia , Humanos , Injeções Intralesionais , Camundongos , Camundongos Nus , Osteossarcoma/metabolismo , Ensaio de Cápsula Sub-Renal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA