Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Molecules ; 26(4)2021 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-33572353

RESUMO

The cholecystokinin-2 receptor (CCK-2R) is overexpressed in several human cancers but displays limited expression in normal tissues. For this reason, it is a suitable target for developing specific radiotracers. In this study, a nastorazepide-based ligand functionalized with a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelator (IP-001) was synthesized and labelled with indium-111. The radiolabeling process yielded >95% with a molar activity of 10 MBq/nmol and a radiochemical purity of >98%. Stability studies have shown a remarkable resistance to degradation (>93%) within 120 h of incubation in human blood. The in vitro uptake of [111In]In-IP-001 was assessed for up to 24 h on a high CCK-2R-expressing tumor cell line (A549) showing maximal accumulation after 4 h of incubation. Biodistribution and single photon emission tomography (SPECT)/CT imaging were evaluated on BALB/c nude mice bearing A549 xenograft tumors. Implanted tumors could be clearly visualized after only 4 h post injection (2.36 ± 0.26% ID/cc), although a high amount of radiotracer was also found in the liver, kidneys, and spleen (8.25 ± 2.21%, 6.99 ± 0.97%, and 3.88 ± 0.36% ID/cc, respectively). Clearance was slow by both hepatobiliary and renal excretion. Tumor retention persisted for up to 24 h, with the tumor to organs ratio increasing over-time and ending with a tumor uptake (1.52 ± 0.71% ID/cc) comparable to liver and kidneys.


Assuntos
Benzodiazepinas/química , Radioisótopos de Índio/farmacocinética , Neoplasias Pulmonares/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Receptor de Colecistocinina B/antagonistas & inibidores , Animais , Apoptose , Proliferação de Células , Feminino , Humanos , Radioisótopos de Índio/administração & dosagem , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/radioterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Compostos Radiofarmacêuticos/administração & dosagem , Receptor de Colecistocinina B/metabolismo , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Theranostics ; 10(25): 11359-11375, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33052220

RESUMO

This is the initial report of an α-based pre-targeted radioimmunotherapy (PRIT) using 225Ac and its theranostic pair, 111In. We call our novel tumor-targeting DOTA-hapten PRIT system "proteus-DOTA" or "Pr." Herein we report the first results of radiochemistry development, radiopharmacology, and stoichiometry of tumor antigen binding, including the role of specific activity, anti-tumor efficacy, and normal tissue toxicity with the Pr-PRIT approach (as α-DOTA-PRIT). A series of α-DOTA-PRIT therapy studies were performed in three solid human cancer xenograft models of colorectal cancer (GPA33), breast cancer (HER2), and neuroblastoma (GD2), including evaluation of chronic toxicity at ~20 weeks of select survivors. Methods: Preliminary biodistribution experiments in SW1222 tumor-bearing mice revealed that 225Ac could not be efficiently pretargeted with current DOTA-Bn hapten utilized for 177Lu or 90Y, leading to poor tumor uptake in vivo. Therefore, we synthesized Pr consisting of an empty DOTA-chelate for 225Ac, tethered via a short polyethylene glycol linker to a lutetium-complexed DOTA for picomolar anti-DOTA chelate single-chain variable fragment (scFv) binding. Pr was radiolabeled with 225Ac and its imaging surrogate, 111In. In vitro studies verified anti-DOTA scFv recognition of [225Ac]Pr, and in vivo biodistribution and clearance studies were performed to evaluate hapten suitability and in vivo targeting efficiency. Results: Intravenously (i.v.) administered 225Ac- or 111In-radiolabeled Pr in mice showed rapid renal clearance and minimal normal tissue retention. In vivo pretargeting studies show high tumor accumulation of Pr (16.71 ± 5.11 %IA/g or 13.19 ± 3.88 %IA/g at 24 h p.i. for [225Ac]Pr and [111In]Pr, respectively) and relatively low uptake in normal tissues (all average ≤ 1.4 %IA/g at 24 h p.i.). Maximum tolerated dose (MTD) was not reached for either [225Ac]Pr alone or pretargeted [225Ac]Pr at administered activities up to 296 kBq/mouse. Single-cycle treatment consisting of α-DOTA-PRIT with either huA33-C825 bispecific anti-tumor/anti-DOTA-hapten antibody (BsAb), anti-HER2-C825 BsAb, or hu3F8-C825 BsAb for targeting GPA33, HER2, or GD2, respectively, was highly effective. In the GPA33 model, no complete responses (CRs) were observed but prolonged overall survival of treated animals was 42 d for α-DOTA-PRIT vs. 25 d for [225Ac]Pr only (P < 0.0001); for GD2, CRs (7/7, 100%) and histologic cures (4/7, 57%); and for HER2, CRs (7/19, 37%) and histologic cures (10/19, 56%) with no acute or chronic toxicity. Conclusions: [225Ac]Pr and its imaging biomarker [111In]Pr demonstrate optimal radiopharmacologic behavior for theranostic applications of α-DOTA-PRIT. For this initial evaluation of efficacy and toxicity, single-cycle treatment regimens were performed in all three systems. Histologic toxicity was not observed, so MTD was not observed. Prolonged overall survival, CRs, and histologic cures were observed in treated animals. In comparison to RIT with anti-tumor IgG antibodies, [225Ac]Pr has a much improved safety profile. Ultimately, these data will be used to guide clinical development of toxicity and efficacy studies of [225Ac]Pr, with the goal of delivering massive lethal doses of radiation to achieve a high probability of cure without toxicity.


Assuntos
Partículas alfa/uso terapêutico , Neoplasias/terapia , Radioimunoterapia/métodos , Compostos Radiofarmacêuticos/administração & dosagem , Nanomedicina Teranóstica/métodos , Actínio/administração & dosagem , Actínio/farmacocinética , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Feminino , Meia-Vida , Compostos Heterocíclicos com 1 Anel/administração & dosagem , Compostos Heterocíclicos com 1 Anel/química , Compostos Heterocíclicos com 1 Anel/farmacocinética , Humanos , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/farmacocinética , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/química , Neoplasias/diagnóstico , Neoplasias/imunologia , Neoplasias/patologia , Radioimunoterapia/efeitos adversos , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Dosagem Radioterapêutica , Distribuição Tecidual , Testes de Toxicidade Crônica , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Nanoscale ; 12(31): 16570-16585, 2020 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-32749427

RESUMO

Stem cells have been utilised as anti-cancer agents due to their ability to home to and integrate within tumours. Methods to augment stem cell homing to tumours are being investigated with the goal of enhancing treatment efficacy. However, it is currently not possible to evaluate both cell localisation and cell viability after engraftment, hindering optimisation of therapy. In this study, luciferase-expressing human adipocyte-derived stem cells (ADSCs) were incubated with Indium-111 radiolabelled iron oxide nanoparticles to produce cells with tri-modal imaging capabilities. ADSCs were administered intravenously (IV) or intracardially (IC) to mice bearing orthotopic breast tumours. Cell fate was monitored using bioluminescence imaging (BLI) as a measure of cell viability, magnetic resonance imaging (MRI) for cell localisation and single photon emission computer tomography (SPECT) for cell quantification. Serial monitoring with multi-modal imaging showed the presence of viable ADSCs within tumours as early as 1-hour post IC injection and the percentage of ADSCs within tumours to be 2-fold higher after IC than IV. Finally, histological analysis was used to validate engraftment of ADSC within tumour tissue. These findings demonstrate that multi-modal imaging can be used to evaluate the efficiency of stem cell delivery to tumours and that IC cell administration is more effective for tumour targeting.


Assuntos
Neoplasias Mamárias Experimentais/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Imagem Multimodal/métodos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Proliferação de Células , Sobrevivência Celular , Rastreamento de Células , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/química , Luciferases/genética , Luciferases/metabolismo , Nanopartículas Magnéticas de Óxido de Ferro/administração & dosagem , Nanopartículas Magnéticas de Óxido de Ferro/química , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Neoplasias Mamárias Experimentais/patologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos
4.
Clin Cancer Res ; 26(22): 5830-5842, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-32816889

RESUMO

PURPOSE: 90Y-FF-21101 is an Yttrium-90-conjugated, chimeric mAb that is highly specific for binding to human placental (P)-cadherin, a cell-to-cell adhesion molecule overexpressed and associated with cancer invasion and metastatic dissemination in many cancer types. We report the clinical activity of 90Y-FF-21101 in a first-in-human phase I study in patients with advanced solid tumors. PATIENTS AND METHODS: The safety and efficacy of 90Y-FF-21101 were evaluated in a phase I 3+3 dose-escalation study in patients with advanced solid tumors (n = 15) over a dose range of 5-25 mCi/m2. Dosimetry using 111In-FF-21101 was performed 1 week prior to assess radiation doses to critical organs. Patients who demonstrated clinical benefit received repeated 90Y-FF-21101 administration every 4 months. RESULTS: 111In-FF-21101 uptake was observed primarily in the spleen, kidneys, testes, lungs, and liver, with tumor uptake observed in the majority of patients. Organ dose estimates for all patients were below applicable limits. P-cadherin expression H-scores ranged from 0 to 242 with 40% of samples exhibiting scores ≥100. FF-21101 protein pharmacokinetics were linear with increasing antibody dose, and the mean half-life was 69.7 (±12.1) hours. Radioactivity clearance paralleled antibody clearance. A complete clinical response was observed in a patient with clear cell ovarian carcinoma, correlating with a high tumor P-cadherin expression. Stable disease was observed in a variety of other tumor types, without dose-limiting toxicity. CONCLUSIONS: The favorable safety profile and initial antitumor activity observed for 90Y-FF-21101 warrant further evaluation of this radioimmunotherapeutic (RIT) approach and provide initial clinical data supporting P-cadherin as a potential target for cancer treatment.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Caderinas/antagonistas & inibidores , Neoplasias/radioterapia , Radioimunoterapia , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/imunologia , Caderinas/genética , Caderinas/imunologia , Antígeno Carcinoembrionário/genética , Adesão Celular/efeitos dos fármacos , Fracionamento da Dose de Radiação , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imunoglobulinas/imunologia , Radioisótopos de Índio/administração & dosagem , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Masculino , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Estadiamento de Neoplasias , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Baço/efeitos dos fármacos , Testículo/efeitos dos fármacos , Radioisótopos de Ítrio/administração & dosagem
5.
Theranostics ; 9(19): 5595-5609, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31534505

RESUMO

Nanomedicines allow active targeting of cancer for diagnostic and therapeutic applications through incorporation of multiple functional components. Frequently, however, clinical translation is hindered by poor intratumoural delivery and distribution. The application of physical stimuli to promote tumour uptake is a viable route to overcome this limitation. In this study, ultrasound-mediated cavitation of microbubbles was investigated as a mean of enhancing the delivery of a liposome designed for chemo-radionuclide therapy targeted to EGFR overexpressing cancer. Method: Liposomes (111In-EGF-LP-Dox) were prepared by encapsulation of doxorubicin (Dox) and surface functionalisation with Indium-111 tagged epidermal growth factor. Human breast cancer cell lines with high and low EGFR expression (MDA-MB-468 and MCF7 respectively) were used to study selectivity of liposomal uptake, subcellular localisation of drug payload, cytotoxicity and DNA damage. Liposome extravasation following ultrasound-induced cavitation of microbubbles (SonoVue®) was studied using a tissue-mimicking phantom. In vivo stability, pharmacokinetic profile and biodistribution were evaluated following intravenous administration of 111In-labelled, EGF-functionalised liposomes to mice bearing subcutaneous MDA-MB-468 xenografts. Finally, the influence of ultrasound-mediated cavitation on the delivery of liposomes into tumours was studied. Results: Liposomes were loaded efficiently with Dox, surface decorated with 111In-EGF and showed selective uptake in MDA-MB-468 cells compared to MCF7. Following binding to EGFR, Dox was released into the intracellular space and 111In-EGF shuttled to the cell nucleus. DNA damage and cell kill were higher in MDA-MB-468 than MCF7 cells. Moreover, Dox and 111In were shown to have an additive cytotoxic effect in MDA-MB-468 cells. US-mediated cavitation increased the extravasation of liposomes in an in vitro gel phantom model. In vivo, the application of ultrasound with microbubbles increased tumour uptake by 66% (p<0.05) despite poor vascularisation of MDA-MB-468 xenografts (as shown by DCE-MRI). Conclusion:111In-EGF-LP-Dox designed for concurrent chemo-radionuclide therapy showed specificity for and cytotoxicity towards EGFR-overexpressing cancer cells. Delivery to tumours was enhanced by the use of ultrasound-mediated cavitation indicating that this approach has the potential to deliver cytotoxic levels of therapeutic radionuclide to solid tumours.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Receptores ErbB/metabolismo , Radioisótopos de Índio/administração & dosagem , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Terapia Combinada , Doxorrubicina/química , Doxorrubicina/farmacocinética , Composição de Medicamentos , Sistemas de Liberação de Medicamentos/instrumentação , Receptores ErbB/genética , Feminino , Humanos , Radioisótopos de Índio/química , Radioisótopos de Índio/farmacocinética , Lipossomos/química , Camundongos , Camundongos Nus , Distribuição Tecidual , Ultrassom
6.
Mol Pharm ; 16(9): 4024-4030, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31345042

RESUMO

OBJECTIVE: Targeting the glucagon-like peptide-1 receptor with radiolabeled exendin is a very promising method to noninvasively determine the ß cell mass in the pancreas, which is needed to unravel the pathophysiology of type 1 and type 2 diabetes. The present study aimed to explore the effects of both hyperglycemia and insulitis on the uptake of exendin in a spontaneous type 1 diabetes mouse model, nonobese diabetic (NOD) mice. METHODS: NOD mice (n = 75, 7-21 weeks old) were injected intravenously with [111In]In-DTPA-exendin-3, and single-photon emission computed tomography (SPECT) images were acquired 1 h pi. The pancreatic accumulation of [111In]In-DTPA-exendin-3 was quantified in vivo using SPECT and by ex vivo counting and correlated to the ß cell mass (BCM). The influence of insulitis and hyperglycemia on the exendin uptake was assessed. RESULTS: The pancreas could be visualized longitudinally using SPECT. A linear correlation was found between the BCM (%) and pancreatic uptake (%ID/g) as measured by ex vivo counting (Pearson r = 0.64, p < 0.001), which was not affected by either insulitis (Pearson r = 0.66, p = 0.83) or hyperglycemia (Pearson r = 0.57, p = 0.51). Biodistribution and ex vivo autoradiography revealed remaining [111In]In-DTPA-exendin-3 uptake in the pancreas despite total ablation of BCM. CONCLUSIONS: Despite hyperglycemia and severe insulitis, we have found a good correlation between BCM and pancreatic exendin uptake, even in a suboptimal model with relatively high background activity.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Hiperglicemia/metabolismo , Células Secretoras de Insulina/metabolismo , Peptídeos/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Autorradiografia , Diabetes Mellitus Tipo 1/diagnóstico por imagem , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/química , Radioisótopos de Índio/metabolismo , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos NOD , Ácido Pentético/administração & dosagem , Ácido Pentético/química , Ácido Pentético/metabolismo , Peptídeos/administração & dosagem , Peptídeos/química , Compostos Radiofarmacêuticos/metabolismo , Distribuição Tecidual
7.
Medicine (Baltimore) ; 98(8): e14625, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30813194

RESUMO

RATIONALE: Adrenal myelolipoma is a benign hormone-inactive tumor composed of hematopoietic tissue and mature adipose tissue. Because this tumor tends to be rich in fat, in many cases it can be diagnosed based on computed tomography (CT) or magnetic resonance imaging (MRI) findings alone. However, in the presence of much necrosis, calcification and hematopoietic tissue and/or the absence of much fat, and in cases with tumor apoplexy, this tumor becomes difficult to differentiate from other tumors. In such cases, a bone marrow scan may be informative as a non-invasive imaging diagnostic method for preoperative diagnosis of the tumor and determination of the method for the surgical treatment. We herein report a case of huge adrenal myelolipoma with the non-adipose portion identified using an Indium chloride (InCl3) bone marrow scan. PATIENT CONCERNS: A 69-year-old woman was referred to our hospital because of a left peritoneal mass detected on a medical checkup. Abdominal CT revealed a mass measuring 14.3 cm in diameter located between the left kidney and the left adrenal gland, which showed coexistence of fat and soft tissue densities. DIAGNOSES: A bone marrow scan is a nuclear medicine examination to assess hematopoietic activity. To avoid excessive resection of the tumor, we thought that a bone marrow scan could be applied for differentiation between myelolipoma and retroperitoneal liposarcoma by evaluating the hematopoietic activity of the tumor. Tumor enucleation was performed, and pathological examination showed a diagnosis of adrenal myelolipoma. INTERVENTION: The patient was treated with laparoscopic enucleation. OUTCOMES: No metastatic recurrence was found during 8 months of follow-up. LESSONS: Diagnosis of myelolipoma by CT and MRI becomes difficult in the presence of a high volume of hematopoietic tissue. In such cases, a bone marrow scan may be informative as a non-invasive imaging diagnostic method for preoperative diagnosis of the tumor and determination of the method of surgical treatment.


Assuntos
Neoplasias das Glândulas Suprarrenais/diagnóstico por imagem , Medula Óssea/diagnóstico por imagem , Mielolipoma/diagnóstico por imagem , Neoplasias das Glândulas Suprarrenais/cirurgia , Idoso , Diagnóstico Diferencial , Feminino , Humanos , Radioisótopos de Índio/administração & dosagem , Laparoscopia/métodos , Mielolipoma/cirurgia , Cintilografia/métodos , Tomografia Computadorizada por Raios X
8.
Oncol Rep ; 41(4): 2371-2378, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30816521

RESUMO

Tissue factor (TF) has emerged as a critical factor in oncogenic events, leading to the development of TF­targeted diagnostic and therapeutic approaches. A non­invasive imaging method to evaluate target molecule expression with high sensitivity and high quantitative ability is imperative for selecting the appropriate patients for TF­targeted therapy. To elucidate the potential of 111In­labeled anti­TF antibody 1849 (111In­1849) as an immuno­single photon emission computed tomography (SPECT) probe targeting TF, we evaluated TF­dependent in vitro binding as well as in vivo biodistribution and tumor accumulation of 111In­1849 in pancreatic cancer cells/models with varying TF expression levels. TF expression levels in five human pancreatic cancer cell lines, BxPC­3, BxPC­3­TF­knockout (BxPC­3­TFKO), Capan­1, PSN­1 and SUIT­2, were examined by immunofluorescence. Binding of 111In­1849 to each cell line was assessed. Biodistribution and imaging studies were also conducted in tumor­bearing mice. Furthermore, the relationship of TF expression with cell binding and tumor uptake was analyzed. In the immunofluorescence studies, BxPC­3 exhibited the highest TF expression, followed by Capan­1, PSN­1, SUIT­2 and BxPC­3­TFKO. Cell binding assays revealed that BxPC­3 cells had the highest 111In­1849 binding, followed by PSN­1, Capan­1 and SUIT­2; no binding was detected in BxPC­3­TFKO cells. The BxPC­3 xenograft was clearly visualized on 111In­1849 SPECT/CT, and the highest uptake was detected on day 4. The biodistribution of 111In­1849 on day 4 revealed that tumor uptake ranged from 8.68 to 50.58% of the injected dose per gram of tissue; BxPC­3 had the highest uptake and SUIT­2 had the lowest. TF expression was significantly associated with cell binding (R2=0.79, P<0.05) and tumor uptake (R2=0.92, P<0.01). The association of 111In­1849 uptake with TF expression suggests the potential application of non­invasive imaging with radiolabelled 1849 for selecting the appropriate patients who would likely respond to TF­targeted therapies in clinical practice.


Assuntos
Imunoconjugados/administração & dosagem , Neoplasias Pancreáticas/diagnóstico por imagem , Tromboplastina/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacocinética , Linhagem Celular Tumoral , Humanos , Imunoconjugados/metabolismo , Imunoconjugados/farmacocinética , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Imagem Molecular/métodos , Neoplasias Pancreáticas/patologia , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Mol Pharm ; 15(7): 2674-2683, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29865791

RESUMO

Radionuclide molecular imaging is a promising tool for visualization of cancer associated molecular abnormalities in vivo and stratification of patients for specific therapies. ADAPT is a new type of small engineered proteins based on the scaffold of an albumin binding domain of protein G. ADAPTs have been utilized to select and develop high affinity binders to different proteinaceous targets. ADAPT6 binds to human epidermal growth factor 2 (HER2) with low nanomolar affinity and can be used for its in vivo visualization. Molecular design of 111In-labeled anti-HER2 ADAPT has been optimized in several earlier studies. In this study, we made a direct comparison of two of the most promising variants, having either a DEAVDANS or a (HE)3DANS sequence at the N-terminus, conjugated with a maleimido derivative of DOTA to a GSSC amino acids sequence at the C-terminus. The variants (designated DOTA-C59-DEAVDANS-ADAPT6-GSSC and DOTA-C61-(HE)3DANS-ADAPT6-GSSC) were stably labeled with 111In for SPECT and 68Ga for PET. Biodistribution of labeled ADAPT variants was evaluated in nude mice bearing human tumor xenografts with different levels of HER2 expression. Both variants enabled clear discrimination between tumors with high and low levels of HER2 expression. 111In-labeled ADAPT6 derivatives provided higher tumor-to-organ ratios compared to 68Ga-labeled counterparts. The best performing variant was DOTA-C61-(HE)3DANS-ADAPT6-GSSC, which provided tumor-to-blood ratios of 208 ± 36 and 109 ± 17 at 3 h for 111In and 68Ga labels, respectively.


Assuntos
Proteínas de Bactérias/química , Desenho de Fármacos , Imagem Molecular/métodos , Neoplasias/diagnóstico por imagem , Receptor ErbB-2/metabolismo , Animais , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular Tumoral , Feminino , Radioisótopos de Gálio/administração & dosagem , Radioisótopos de Gálio/química , Radioisótopos de Gálio/metabolismo , Humanos , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/química , Radioisótopos de Índio/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Engenharia de Proteínas , Cintilografia/métodos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Mol Pharm ; 15(3): 1150-1159, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29314858

RESUMO

A metal-chelating polymer (MCP) with a polyglutamide (PGlu) backbone presenting on average 13 DOTA (tetraazacyclododecane-1,4,7,10-tetraacetic acid) chelators for complexing 111In or 177Lu and 10 polyethylene glycol (PEG) chains to minimize liver and spleen uptake was conjugated to antiepidermal growth factor receptor (EGFR) monoclonal antibody (mAb), panitumumab. Because panitumumab-MCP may be dual-labeled with 111In and 177Lu for SPECT, or radioimmunotherapy (RIT) exploiting the Auger electrons or ß-particle emissions, respectively, we propose that panitumumab-MCP could be a useful theranostic agent for EGFR-positive PnCa. Bioconjugation was achieved by reaction of a hydrazine nicotinamide (HyNIC) group on the MCP with an aryl aromatic aldehyde introduced into panitumumab by reaction with succinimidyl-4-formylbenzamide (S-4FB). The conjugation reaction was monitored by measurement of the chromophoric bis-aryl hydrazone bond formed (ε350 nm = 24 500 M-1 cm-1) to achieve two MCPs/panitumumab. Labeling of panitumumab-MCP with 111In or 177Lu demonstrated that masses as small as 0.1 µg were labeled to >90% labeling efficiency (L.E.) and a specific activity (SA) of >70 MBq/µg. Panitumumab-DOTA incorporating two DOTA per mAb was labeled with 111In or 177Lu to a maximum SA of 65 MBq/µg and 46 MBq/µg, respectively. Panitumumab-MCP-177Lu exhibited saturable binding to EGFR-overexpressing MDA-MB-468 human breast cancer cells. The Kd for binding of panitumumab-MCP-177Lu to EGFR (2.2 ± 0.6 nmol/L) was not significantly different than panitumumab-DOTA-177Lu (1.0 ± 0.4 nmol/L). 111In and 177Lu were stably complexed to panitumumab-MCP. Panitumumab-MCP-111In exhibited similar whole body retention (55-60%) as panitumumab-DOTA-111In in NOD-scid mice up to 72 h postinjection (p.i.) and equivalent excretion of radioactivity into the urine and feces. The uptake of panitumumab-MCP-111In in most normal tissues in NOD-scid mice with EGFR-positive PANC-1 human pancreatic cancer (PnCa) xenografts at 72 h p.i. was not significantly different than panitumumab-DOTA-111In, except for the liver which was 3-fold greater for panitumumab-MCP-111In. Tumor uptake of panitumumab-MCP-111In (6.9 ± 1.3%ID/g) was not significantly different than panitumumab-DOTA-11In (6.6 ± 3.3%ID/g). Tumor uptake of panitumumab-MCP-111In and panitumumab-DOTA-111In were reduced by preadministration of excess panitumumab, suggesting EGFR-mediated uptake. Tumor uptake of nonspecific IgG-MCP (5.4 ± 0.3%ID/g) was unexpectedly similar to panitumumab-MCP-111In. An increased hydrodynamic radius of IgG when conjugated to an MCP may encourage tumor uptake via the enhanced permeability and retention (EPR) effect. Tumor uptake of panitumumab-DOTA-111In was 3.5-fold significantly higher than IgG-DOTA-111In. PANC-1 tumors were imaged by microSPECT/CT at 72 h p.i. of panitumumab-MCP-111In or panitumumab-DOTA-111In. Tumors were not visualized with preadministration of excess panitumumab to block EGFR, or with nonspecific IgG radioimmunoconjugates. We conclude that linking panitumumab to an MCP enabled higher SA labeling with 111In and 177Lu than DOTA-conjugated panitumumab, with preserved EGFR binding in vitro and comparable tumor localization in vivo in mice with s.c. PANC-1 human PnCa xenografts. Normal tissue distribution was similar except for the liver which was higher for the polymer radioimmunoconjugates.


Assuntos
Imunoconjugados/administração & dosagem , Neoplasias Pancreáticas/radioterapia , Panitumumabe/administração & dosagem , Radioimunoterapia/métodos , Nanomedicina Teranóstica/métodos , Animais , Linhagem Celular Tumoral , Quelantes/química , Receptores ErbB/antagonistas & inibidores , Feminino , Compostos Heterocíclicos com 1 Anel/química , Humanos , Imunoconjugados/química , Imunoconjugados/farmacocinética , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/química , Lutécio/administração & dosagem , Lutécio/química , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Panitumumabe/química , Panitumumabe/farmacocinética , Polietilenoglicóis/química , Radioisótopos/administração & dosagem , Radioisótopos/química , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Curr Opin Urol ; 28(2): 191-196, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29028767

RESUMO

PURPOSE OF REVIEW: With Gallium (Ga)-prostate-specific membrane antigen (PSMA) PET/computed tomography (CT) as emerging imaging technique offering superior detection rates in biochemical recurrent prostate cancer, salvage lymph node dissection has gained increasing interest in localized oligometastatic prostate cancer. Currently, PSMA-targeting small molecules cannot only be linked to positron-emitting isotopes for imaging but also be labelled with γ-radiation emitting isotopes. These modified PSMA agents are evaluated for intraoperative guidance for resection of metastatic lymph nodes. This review aims to review current knowledge on the novel technique of PSMA-radioguided surgery. RECENT FINDINGS: Recently radiolabeling of PSMA ligands with Indium and Technetium as γ emitter has been established. After preoperative intravenous injection of these novel PSMA agents single photon emission computed tomography/CT imaging can be performed using the γ-emitting properties. Although its diagnostic performance seems to be inferior to Ga-PSMA PET/CT, intraoperative guidance by the use of a γ probe was reported to facilitate detection and resection of tumour-infiltrated soft tissue. First follow-up data suggests favourable outcomes concerning prostate-specific antigen progression and treatment-free survival in a subset of patients. SUMMARY: Although current knowledge is still limited and published data is sparse salvage surgery in recurrent prostate cancer facilitated by γ-emitting PSMA agents seems feasible. However, careful identification of ideal candidates for those procedures is mandatory.


Assuntos
Metástase Linfática/diagnóstico por imagem , Imagem Molecular/métodos , Recidiva Local de Neoplasia/cirurgia , Neoplasias da Próstata/cirurgia , Terapia de Salvação/métodos , Antígenos de Superfície/metabolismo , Estudos de Viabilidade , Radioisótopos de Gálio/administração & dosagem , Glutamato Carboxipeptidase II/metabolismo , Humanos , Radioisótopos de Índio/administração & dosagem , Ligantes , Excisão de Linfonodo/métodos , Linfonodos/diagnóstico por imagem , Linfonodos/patologia , Linfonodos/cirurgia , Metástase Linfática/patologia , Masculino , Recidiva Local de Neoplasia/diagnóstico por imagem , Recidiva Local de Neoplasia/patologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/patologia , Compostos Radiofarmacêuticos/administração & dosagem , Tecnécio/administração & dosagem
12.
Sci Rep ; 7(1): 12341, 2017 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-28951589

RESUMO

Nuclear medicine examinations for imaging gliomas have been introduced into clinical practice to evaluate the grade of malignancy and determine sampling locations for biopsies. However, these modalities have some limitations. Tissue factor (TF) is overexpressed in various types of cancers, including gliomas. We thus generated an anti-human TF monoclonal antibody (mAb) clone 1849. In the present study, immunohistochemistry performed on glioma specimens using anti-TF 1849 mAb showed that TF expression in gliomas increased in proportion to the grade of malignancy based on the World Health Organization (WHO) classification, and TF was remarkably expressed in necrosis and pseudopalisading cells, the histopathological hallmarks of glioblastoma multiforme (GBM). Furthermore, in both fluorescence and single-photon emission computed tomography/computed tomography (SPECT/CT) imaging studies, anti-TF 1849 IgG efficiently accumulated in TF-overexpressing intracranial tumours in mice. Although further investigation is required for a future clinical use of immuno-SPECT with 111In-labelled anti-TF 1849 IgG, the immuno-SPECT may represent a unique imaging modality that can visualize the biological characteristics of gliomas differently from those obtained using the existing imaging modalities and may be useful to evaluate the grade of malignancy and determine sampling locations for biopsies in patients with glioma, particularly GBM.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Glioma/diagnóstico por imagem , Imunoconjugados/administração & dosagem , Imagem Molecular/métodos , Tromboplastina/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Feminino , Glioma/patologia , Humanos , Imunoconjugados/química , Imunoconjugados/imunologia , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Tromboplastina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Mol Pharm ; 14(10): 3457-3463, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28826214

RESUMO

Complete resection of tumor lesions in advanced stage ovarian cancer patients is of utmost importance, since the extent of residual disease after surgery strongly affects survival. Intraoperative imaging may be useful to improve surgery in these patients. Farletuzumab is a humanized IgG1 antibody that specifically recognizes the folate receptor alpha (FRα). Labeled with a radiolabel and a fluorescent dye, farletuzumab may be used for the intraoperative detection of ovarian cancer lesions. The current aim is to demonstrate the feasibility of FRα-targeted dual-modality imaging using 111In-farletuzumab-IRDye800CW in an intraperitoneal ovarian cancer model. Biodistribution studies were performed 3 days after injection of 3, 10, 30, or 100 µg of 111In-farletuzumab-IRDye800CW in mice with subcutaneous IGROV-1 tumors (5 mice per group). In mice with intraperitoneal IGROV-1 tumors the nonspecific uptake of 111In-farletuzumab-IRDye800CW was determined by coinjecting an excess of unlabeled farletuzumab. MicroSPECT/CT and fluorescence imaging were performed 3 days after injection of 10 µg of 111In-farletuzumab-IRDye800CW. FRα expression in tumors was determined immunohistochemically. Optimal tumor-to-blood-ratios (3.4-3.7) were obtained at protein doses up to 30 µg. Multiple intra-abdominal tumor lesions were clearly visualized by microSPECT/CT, while uptake in normal tissues was limited. Fluorescence imaging was used to visualize and guide resection of superficial tumors. Coinjection of an excess of unlabeled farletuzumab significantly decreased tumor uptake of 111In-farletuzumab-IRDye800CW (69.4 ± 27.6 versus 18.3 ± 2.2% ID/g, p < 0.05). Immunohistochemical analyses demonstrated that the radioactive and fluorescent signal corresponded with FRα-expressing tumor lesions. FRα-targeted SPECT/fluorescence imaging using 111In-farletuzumab-IRDye800CW can be used to detect ovarian cancer in vivo and could be a valuable tool for enhanced intraoperative tumor visualization in patients with intraperitoneal metastases of ovarian cancer.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Receptor 1 de Folato/antagonistas & inibidores , Cuidados Intraoperatórios/métodos , Neoplasias Ovarianas/diagnóstico por imagem , Cirurgia Assistida por Computador/métodos , Animais , Anticorpos Monoclonais Humanizados/química , Benzenossulfonatos/administração & dosagem , Benzenossulfonatos/química , Linhagem Celular Tumoral , Estudos de Viabilidade , Feminino , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/química , Receptor 1 de Folato/imunologia , Humanos , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/química , Indóis/administração & dosagem , Indóis/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Imagem Molecular/métodos , Imagem Óptica/métodos , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/cirurgia , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Dokl Biochem Biophys ; 473(1): 85-87, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28510138

RESUMO

A new modular nanotransporter (MNT) for the delivery of anticancer agents into the nuclei of cells with folate receptor overexpression was created. An effective method for acceding labeling of this MNT with Auger electron emitter 111In has been developed. A significant therapeutic effect was observed after a single intratumoral injection of the new 111In-labeled MNT to mice grafted with human cervical carcinoma characterized by folate receptor overexpression.


Assuntos
Núcleo Celular/metabolismo , Elétrons , Transportadores de Ácido Fólico/metabolismo , Regulação Neoplásica da Expressão Gênica , Radioisótopos de Índio/metabolismo , Radioisótopos de Índio/uso terapêutico , Animais , Células HeLa , Humanos , Radioisótopos de Índio/administração & dosagem , Camundongos
15.
J Control Release ; 256: 1-8, 2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28412225

RESUMO

High Intensity Focused Ultrasound (HIFU) is an emerging noninvasive, nonionizing physical energy based modality to ablate solid tumors with high power, or increase local permeability in tissues/tumors in pulsed mode with relatively low power. Compared with traditional ablative HIFU, nondestructive pulsed HIFU (pHIFU) is present in the majority of novel applications recently developed for enhancing the delivery of drugs and genes. Previous studies have demonstrated the capability of pHIFU to change tissue local permeability for enhanced drug delivery in both mouse tumors and mouse muscle. Further study based on bulk tissues in large animals and clinical HIFU system revealed correlation between therapeutic effect and thermal parameters, which was absent in the previous mouse studies. In this study, we further investigated the relation between the therapeutic effect of pHIFU and thermal parameters in bulky normal muscle tissues based on a rabbit model and a preclinical HIFU system. Correlation between therapeutic effect and thermal parameters was confirmed in our study on the same bulk tissues although different HIFU systems were used. Following the study in bulky normal muscle tissues, we further created bulky tumor model with VX2 tumors implanted on both hind limbs of rabbits and investigated the feasibility to enhance tumor permeability in bulky VX2 tumors in a rabbit model using pHIFU technique. A radiolabeled peptidomimetic integrin antagonist, 111In-DOTA-IA, was used following pHIFU treatment in our study to target VX2 tumor and serve as the radiotracer for follow-up single-photon emission computed tomography (SPECT) scanning. The results have shown significantly elevated uptake of 111In-DOTA-IA in the area of VX2 tumors pretreated by pHIFU compared with the control VX2 tumors not being pretreated by pHIFU, and statistical analysis revealed averaged 34.5% enhancement 24h after systematic delivery of 111In-DOTA-IA in VX2 tumors pretreated by pHIFU compared with the control VX2 tumors.


Assuntos
Complexos de Coordenação/administração & dosagem , Sistemas de Liberação de Medicamentos , Compostos Heterocíclicos com 1 Anel/administração & dosagem , Ablação por Ultrassom Focalizado de Alta Intensidade , Radioisótopos de Índio/administração & dosagem , Neoplasias Musculares , Animais , Nádegas/diagnóstico por imagem , Complexos de Coordenação/farmacocinética , Complexos de Coordenação/uso terapêutico , Feminino , Compostos Heterocíclicos com 1 Anel/farmacocinética , Compostos Heterocíclicos com 1 Anel/uso terapêutico , Radioisótopos de Índio/farmacocinética , Radioisótopos de Índio/uso terapêutico , Imageamento por Ressonância Magnética , Neoplasias Musculares/diagnóstico por imagem , Neoplasias Musculares/metabolismo , Neoplasias Musculares/terapia , Permeabilidade , Coelhos , Tomografia Computadorizada de Emissão de Fóton Único
16.
Bioorg Med Chem ; 25(4): 1406-1412, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28089587

RESUMO

Insulinoma is a tumor derived from pancreatic ß-cells, and the resulting hyperinsulinemia leads to characteristic hypoglycemia. Recent studies have reported the frequent overexpression of glucagon-like peptide-1 receptor (GLP-1R) in human insulinomas, suggesting that the binding of a radiolabeled compound to GLP-1R is useful for the imaging of such tumors. Exendin(9-39), a fragment peptide of exendin-3 and -4, binds GLP-1R with high affinity and acts as an antagonist. Accordingly, radiolabeled exendin(9-39) derivatives have also been investigated as insulinoma imaging probes that might be less likely to induce hypoglycemia. In this study, we synthesized a novel indium-111 (111In)-benzyl-diethylenetriaminepentaacetic acid (111In-BnDTPA)-conjugated exendin(9-39), 111In-BnDTPA-exendin(9-39), and evaluated its utility as a probe for the SPECT imaging of insulinoma. natIn-BnDTPA-exendin(9-39) exhibited a high affinity for GLP-1R (IC50=2.5nM), stability in plasma, and a specific activity that improved following reactions with a solvent and solubilizer. Regarding the in vivo biodistribution of 111In-BnDTPA-exendin(9-39) in INS-1 tumor-bearing mice, high uptake levels were observed in tumors (14.6%ID/g at 15min), with corresponding high tumor-to-blood (T/B), tumor-to-muscle (T/M), and tumor-to-pancreas (T/P) ratios (T/B=2.55, T/M=22.7, T/P=2.7 at 1h). The pre-administration of excess nonradioactive exendin(9-39) significantly reduced accumulation in both the tumor and pancreas (76% and 68% inhibition, respectively) at 1h after 111In-BnDTPA-exendin(9-39) injection, indicating that the GLP-1R mediated a majority of 111In-BnDTPA-exendin(9-39) uptake in the tumor and pancreas. Finally, 111In-BnDTPA-exendin(9-39) SPECT/CT studies in mice yielded clear images of tumors at 30min post-injection. These results suggest that 111In-BnDTPA-exendin(9-39) could be a useful SPECT molecular imaging probe for the detection and exact localization of insulinomas.


Assuntos
Radioisótopos de Índio/química , Insulinoma/diagnóstico por imagem , Imagem Molecular , Neoplasias Pancreáticas/diagnóstico por imagem , Fragmentos de Peptídeos/química , Compostos Radiofarmacêuticos/química , Tomografia Computadorizada de Emissão de Fóton Único , Animais , Relação Dose-Resposta a Droga , Radioisótopos de Índio/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Estrutura Molecular , Neoplasias Experimentais/diagnóstico por imagem , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/síntese química , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/síntese química , Ratos , Relação Estrutura-Atividade , Células Tumorais Cultivadas
17.
Cancer Biother Radiopharm ; 31(9): 324-329, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27831759

RESUMO

By using radio-labeled multifunctional superparamagnetic iron oxide nanoparticles (SPIONs) and an alternating magnetic field (AMF), we carried out targeted hyperthermia, drug delivery, radio-immunotherapy (RIT), and controlled chemotherapy of cancer tumors. We synthesized and characterized Indium-111-labeled, Trastuzumab and Doxorubicin (DOX)-conjugated APTES-PEG-coated SPIONs in our previous work. Then, we evaluated their capability in SPECT/MRI (single photon emission computed tomography/magnetic resonance imaging) dual modal molecular imaging, targeting, and controlled release. In this research, AMF was introduced to evaluate therapeutic effects of magnetic hyperthermia on radionuclide-chemo therapy of HER2+ cells and tumor (HER2+)-bearing mice. In vitro and in vivo experiments using synthesized complex were repeated under an AMF (f: 100 KHz, H: 280 Gs). Instead of an intra-tumor injection in most hyperthermia experiments, SPIONs were injected to the tail vein, based on our delivery strategies. For magnetic delivery, we held a permanent Nd-B-Fe magnet near the tumor region. The results showed that simultaneous magnetic hyperthermia enhanced SKBR3 cancer cells, killing by 24%, 28%, 33%, and 80% at 48 hours post-treatment for treated cells with (1) bare SPIONs; (2) antibody-conjugated, DOX-free, surface-modified SPIONs; (3) 111In-labeled, antibody-conjugated surface-modified SPIONs; and (4) 111In-labeled, antibody- and DOX-conjugated surface-modified SPIONs, respectively. Moreover, tumor volume inhibitory rate was 85% after a 28 day period of treatment. By using this method, multimodal imaging-guided, targeted hyperthermia, RIT, and controlled chemotherapy could be achievable in the near future.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/terapia , Compostos Férricos/administração & dosagem , Febre/terapia , Magnetoterapia/métodos , Nanopartículas/administração & dosagem , Receptor ErbB-2/biossíntese , Animais , Linhagem Celular Tumoral , Terapia Combinada , Doxorrubicina/administração & dosagem , Doxorrubicina/química , Feminino , Humanos , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/química , Magnetismo/métodos , Camundongos , Camundongos Endogâmicos BALB C , Radioimunoterapia/métodos , Distribuição Aleatória , Trastuzumab/administração & dosagem , Trastuzumab/química , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Int J Radiat Biol ; 92(11): 716-723, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26999580

RESUMO

Purpose Radiolabeled antibodies and peptides hold promise for molecular radiotherapy but are often limited by a low payload resulting in inadequate delivery of radioactivity to tumour tissue and, therefore, modest therapeutic effect. We developed a facile synthetic method of radiolabeling indium-111 (111In) to epidermal growth factor (EGF)-gold nanoparticles (111In-EGF-Au NP) with a high payload. Materials and methods EGF-Au NP were prepared via an interaction between gold and the disulphide bonds of EGF and radiolabeled using 111InCl3. Targeting efficiency was investigated by quantitating internalized radioactivity and by confocal imaging following exposure of MDA-MB-468 (1.3 × 106 EGFR/cell) and MCF-7 (104 EGFR/cell) cells to Cy3-EGF-Au NP. Cytotoxicity was evaluated in clonogenic assays. Results The proportion of total administered radioactivity that was internalized by MDA-MB-468 and MCF-7 cells was 15% and 1.3%, respectively (mixing ratio of EGF:Au of 160). This differential uptake in the two cell lines was confirmed using confocal microscopy. 111In-EGF-Au NP were significantly more radiotoxic to MDA-MB-468 than MCF-7 cells with a surviving fraction of 17.1 ± 4.4% versus 89.8 ± 1.4% (p < 0.001) after exposure for 4 h. Conclusions An 111In-labeled EGF-Au nanosystem was developed. It enabled targeted delivery of a high 111In payload specifically to EGFR-positive cancer cells leading to radiotoxicity that can be exploited for molecularly targeted radiotherapy.


Assuntos
Fator de Crescimento Epidérmico/farmacocinética , Receptores ErbB/metabolismo , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/farmacocinética , Nanopartículas Metálicas/química , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/radioterapia , Sobrevivência Celular/efeitos da radiação , Materiais Revestidos Biocompatíveis , Ouro/química , Humanos , Radioisótopos de Índio/química , Células MCF-7 , Terapia de Alvo Molecular/métodos , Nanocápsulas/química , Neoplasias Experimentais/patologia , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Resultado do Tratamento
19.
J Control Release ; 226: 115-23, 2016 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-26869546

RESUMO

Poly(ethylene glycol) (PEG) is an artificial but biocompatible hydrophilic polymer that has been widely used in clinical products. To evaluate the feasibility of using PEG derivative itself as a tumor imaging carrier via an enhanced permeability and retention (EPR) effect, we prepared indium-111-labeled PEG ((111)In-DTPA-PEG) and indocyanine green (ICG)-labeled PEG (ICG-PEG) with PEG molecular weights of 5-40kDa and investigated their in vivo biodistribution in colon26 tumor-bearing mice. Thereafter, single-photon emission computed tomography (SPECT) and photoacoustic (PA) imaging studies were performed. The in vivo biodistribution studies demonstrated increased tumor uptake and a prolongation of circulation half-life as the molecular weight of PEG increased. Although the observed differences in in vivo biodistribution were dependent on the labeling method ((111)In or ICG), the tumor-to-normal tissue ratios were comparable. Because PEG-based probes with a molecular weight of 20kDa (PEG20) showed a preferable biodistribution (highest accumulation among tissues excised and relatively high tumor-to-blood ratios), an imaging study using (111)In-DTPA-PEG20 and ICG-PEG20 was performed. Colon26 tumors inoculated in the right shoulder were clearly visualized by SPECT 24h after administration. Furthermore, PA imaging using ICG-PEG20 also detected tumor regions, and the detected PA signals increased in proportion with the injected dose. These results suggest that PEG derivatives (20kDa) serve as robust diagnostic drug carriers for tumor imaging.


Assuntos
Corantes/administração & dosagem , Portadores de Fármacos/química , Radioisótopos de Índio/administração & dosagem , Verde de Indocianina/administração & dosagem , Neoplasias/diagnóstico por imagem , Polietilenoglicóis/química , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Animais , Linhagem Celular Tumoral , Corantes/química , Corantes/farmacocinética , Radioisótopos de Índio/química , Radioisótopos de Índio/farmacocinética , Verde de Indocianina/química , Verde de Indocianina/farmacocinética , Camundongos , Ácido Pentético/química , Técnicas Fotoacústicas , Distribuição Tecidual
20.
Mol Pharm ; 13(3): 1158-65, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26883169

RESUMO

Optimal biodistribution and prolonged circulation of nanocarriers improve diagnostic and therapeutic effects of enhanced permeability and retention-based nanomedicines. Despite extensive use of Pluronics in polymer-based pharmaceuticals, the influence of different poly(ethylene oxide) (PEO) block length and aggregation state on the biodistribution of the carriers is rather unexplored. In this work, we studied these effects by evaluating the biodistribution of Pluronic unimers and cross-linked micelles with different PEO block size. In vivo biodistribution of (111)In-radiolabeled Pluronic nanocarriers was investigated in healthy mice using single photon emission computed tomography. All carriers show fast uptake in the organs from the reticuloendothelial system followed by a steady elimination through the hepatobiliary tract and renal filtration. The PEO block length affects the initial renal clearance of the compounds and the overall liver uptake. The aggregation state influences the long-term accumulation of the nanocarriers in the liver. We showed that the circulation time and elimination pathways can be tuned by varying the physicochemical properties of Pluronic copolymers. Our results can be beneficial for the design of future Pluronic-based nanomedicines.


Assuntos
Portadores de Fármacos , Imagem Molecular/métodos , Nanopartículas/química , Poloxâmero/química , Polietilenoglicóis/química , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Tomografia Computadorizada por Raios X/métodos , Animais , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/química , Radioisótopos de Índio/farmacocinética , Camundongos , Camundongos Endogâmicos A , Micelas , Nanopartículas/administração & dosagem , Polímeros/química , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA