Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38940841

RESUMO

PURPOSE: The radionuclide pair cerium-134/lanthanum-134 (134Ce/134La) was recently proposed as a suitable diagnostic counterpart for the therapeutic alpha-emitter actinium-225 (225Ac). The unique properties of 134Ce offer perspectives for developing innovative in vivo investigations that are not possible with 225Ac. In this work, 225Ac- and 134Ce-labelled tracers were directly compared using internalizing and slow-internalizing cancer models to evaluate their in vivo comparability, progeny meandering, and potential as a matched theranostic pair for clinical translation. Despite being an excellent chemical match, 134Ce/134La has limitations to the setting of quantitative positron emission tomography imaging. METHODS: The precursor PSMA-617 and a macropa-based tetrazine-conjugate (mcp-PEG8-Tz) were radiolabelled with 225Ac or 134Ce and compared in vitro and in vivo using standard (radio)chemical methods. Employing biodistribution studies and positron emission tomography (PET) imaging in athymic nude mice, the radiolabelled PSMA-617 tracers were evaluated in a PC3/PIP (PC3 engineered to express a high level of prostate-specific membrane antigen) prostate cancer mouse model. The 225Ac and 134Ce-labelled mcp-PEG8-Tz were investigated in a BxPC-3 pancreatic tumour model harnessing the pretargeting strategy based on a trans-cyclooctene-modified 5B1 monoclonal antibody. RESULTS: In vitro and in vivo studies with both 225Ac and 134Ce-labelled tracers led to comparable results, confirming the matching pharmacokinetics of this theranostic pair. However, PET imaging of the 134Ce-labelled precursors indicated that quantification is highly dependent on tracer internalization due to the redistribution of 134Ce's PET-compatible daughter 134La. Consequently, radiotracers based on internalizing vectors like PSMA-617 are suited for this theranostic pair, while slow-internalizing 225Ac-labelled tracers are not quantitatively represented by 134Ce PET imaging. CONCLUSION: When employing slow-internalizing vectors, 134Ce might not be an ideal match for 225Ac due to the underestimation of tumour uptake caused by the in vivo redistribution of 134La. However, this same characteristic makes it possible to estimate the redistribution of 225Ac's progeny noninvasively. In future studies, this unique PET in vivo generator will further be harnessed to study tracer internalization, trafficking of receptors, and the progression of the tumour microenvironment.

2.
Bioorg Med Chem Lett ; 108: 129803, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38777280

RESUMO

Targeted delivery of radionuclides to tumors is significant in theranostics applications for precision medicine. Pre-targeting, in which a tumor-targeting vehicle and a radionuclide-loaded effector small molecule are administered separately, holds promise since it can reduce unnecessary internal radiation exposure of healthy cells and can minimize radiation decay. The success of the pre-targeting delivery requires an in vivo-stable tumor-targeting vehicle selectively binding to tumor antigens and an in vivo-stable small molecule effector selectively binding to the vehicle accumulated on the tumor. We previously reported a drug delivery system composed of a low-immunogenic streptavidin with weakened affinity to endogenous biotin and a bis-iminobiotin with high affinity to the engineered streptavidin. It was, however, unknown whether the bis-iminobiotin is stable in vivo when administered alone for the pre-targeting applications. Here we report a new in vivo-stable bis-iminobiotin derivative. The keys to success were the identification of the degradation site of the original bis-iminobiotin treated with mouse plasma and the structural modification of the degradation site. We disclosed the successful pre-targeting delivery of astatine-211 (211At), α-particle emitter, to the CEACAM5-positive tumor in xenograft mouse models.


Assuntos
Biotina , Estreptavidina , Animais , Estreptavidina/química , Camundongos , Biotina/química , Humanos , Sistemas de Liberação de Medicamentos , Linhagem Celular Tumoral , Mutação , Estrutura Molecular
3.
J Nanobiotechnology ; 22(1): 101, 2024 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-38462598

RESUMO

BACKGROUND: Radiotheranostics differs from the vast majority of other cancer therapies in its capacity for simultaneous imaging and therapy, and it is becoming more widely implemented. A balance between diagnostic and treatment requirements is essential for achieving effective radiotheranostics. Herein, we propose a proof-of-concept strategy aiming to address the profound differences in the specific requirements of the diagnosis and treatment of radiotheranostics. RESULTS: To validate the concept, we designed an s-tetrazine (Tz) conjugated prostate-specific membrane antigen (PSMA) ligand (DOTA-PSMA-Tz) for 68Ga or 177Lu radiolabeling and tumor radiotheranostics, a trans-cyclooctene (TCO) modified Pd@Au nanoplates (Pd@Au-PEG-TCO) for signal amplification, respectively. We then demonstrated this radiotheranostic strategy in the tumor-bearing mice with the following three-step procedures: (1) i.v. injection of the [68Ga]Ga-PSMA-Tz for diagnosis; (2) i.v. injection of the signal amplification module Pd@Au-PEG-TCO; (3) i.v. injection of the [177Lu]Lu-PSMA-Tz for therapy. Firstly, this strategy was demonstrated in 22Rv1 tumor-bearing mice via positron emission tomography (PET) imaging with [68Ga]Ga-PSMA-Tz. We observed significantly higher tumor uptake (11.5 ± 0.8%ID/g) with the injection of Pd@Au-PEG-TCO than with the injection [68Ga]Ga-PSMA-Tz alone (5.5 ± 0.9%ID/g). Furthermore, we validated this strategy through biodistribution studies of [177Lu]Lu-PSMA-Tz, with the injection of the signal amplification module, approximately five-fold higher tumor uptake of [177Lu]Lu-PSMA-Tz (24.33 ± 2.53% ID/g) was obtained when compared to [177Lu]Lu-PSMA-Tz alone (5.19 ± 0.26%ID/g) at 48 h post-injection. CONCLUSION: In summary, the proposed strategy has the potential to expand the toolbox of pretargeted radiotherapy in the field of theranostics.


Assuntos
Neoplasias Colorretais , Compostos Radiofarmacêuticos , Masculino , Animais , Camundongos , Radioisótopos de Gálio , Distribuição Tecidual , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia
4.
Biochem Biophys Res Commun ; 673: 23-28, 2023 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-37354656

RESUMO

This study aimed to establish the radio-immune imaging protocol on the basis of Avidin/Biotin system. The programmed death-ligand 1 (PD-L1) antibody (Atezolizumab) was employed as the primary molecule in targeting PD-L1, and the two-step strategy, consisting of the first injection of Avidin-conjugated PD-L1 monoclonal antibody (Atezolizumab) and the second injection of 7.4 MBq 68Ga-Biotin with a 60 h interval, was then verified on the colon cancer-bearing mice. PET imaging was performed at 30, 90, 180 min to measure the standard uptake value and tumor to liver ratios. Cellular binding experiments and in vivo distribution showed that the conjugation of Avidin did not affect the affinity of Atezolizumab to PD-L1 antigen. Biotin was radio-labeled with 68Ga with radiolabeling efficiency of 70.5 ± 3.5% and purification was needed to increase the radiochemical purity. For PD-L1-positive tumors, SUVmax was 0.38 ± 0.06 in the Avidin-Atezolizumab pre-treated mice at 90 min; the tumor/liver ratios of pre-targeting group were 1.06 ± 0.19 and 0.97 ± 0.16 at 30 and 90 min, while the absence of pre-treatment of Avidin was of the lower ratios as 0.88 ± 0.01 and 0.54 ± 0.11 when 68Ga-Biotin served as the radiopharmaceutical as well. In conclusion, pre-targeting immunoPET strategy can elevate the target-to-nontarget ratio, decrease the blood background and shorten the interval between injection of radiopharmaceuticals and PET scan, providing a highly PD-L1-specific and sensitive imaging method for the detection of tumorous immune micro-environment.


Assuntos
Biotina , Neoplasias do Colo , Camundongos , Animais , Avidina , Antígeno B7-H1/metabolismo , Radioisótopos de Gálio , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos , Linhagem Celular Tumoral , Microambiente Tumoral
5.
Proc Natl Acad Sci U S A ; 117(45): 28316-28327, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33106429

RESUMO

Over the past decade, theranostic imaging has emerged as a powerful clinical tool in oncology for identifying patients likely to respond to targeted therapies and for monitoring the response of patients to treatment. Herein, we report a theranostic approach to pretargeted radioimmunotherapy (PRIT) based on a pair of radioisotopes of copper: positron-emitting copper-64 (64Cu, t1/2 = 12.7 h) and beta particle-emitting copper-67 (67Cu, t1/2 = 61.8 h). This strategy is predicated on the in vivo ligation between a trans-cyclooctene (TCO)-bearing antibody and a tetrazine (Tz)-based radioligand via the rapid and bioorthogonal inverse electron-demand Diels-Alder reaction. Longitudinal therapy studies were conducted in a murine model of human colorectal carcinoma using an immunoconjugate of the huA33 antibody modified with TCO (huA33-TCO) and a 67Cu-labeled Tz radioligand ([67Cu]Cu-MeCOSar-Tz). The injection of huA33-TCO followed 72 h later by the administration of 18.5, 37.0, or 55.5 MBq of [67Cu]Cu-MeCOSar-Tz produced a dose-dependent therapeutic response, with the median survival time increasing from 68 d for the lowest dose to >200 d for the highest. Furthermore, we observed that mice that received the highest dose of [67Cu]Cu-MeCOSar-Tz in a fractionated manner exhibited improved hematological values without sacrificing therapeutic efficacy. Dual radionuclide experiments in which a single administration of huA33-TCO was followed by separate injections of [64Cu]Cu-MeCOSar-Tz and [67Cu]Cu-MeCOSar-Tz revealed that the positron emission tomography images produced by the former accurately predicted the efficacy of the latter. In these experiments, a correlation was observed between the tumoral uptake of [64Cu]Cu-MeCOSar-Tz and the subsequent therapeutic response to [67Cu]Cu-MeCOSar-Tz.


Assuntos
Radioisótopos de Cobre/farmacologia , Radioisótopos de Cobre/uso terapêutico , Medicina de Precisão/métodos , Radioimunoterapia/métodos , Animais , Anticorpos , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Reação de Cicloadição , Relação Dose-Resposta a Droga , Feminino , Humanos , Imunoconjugados , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons/métodos , Radioisótopos/farmacologia , Radioisótopos/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Labelled Comp Radiopharm ; 66(1): 22-30, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36539610

RESUMO

Pretargeting imaging has gained a lot of prominence, due to its excellent bioorthogonality and improved imaging contrast compared to conventional imaging. A new iodo tetrazine (Tz) derivative has been synthesized and further developed into the corresponding iodine-125 (125 I) analog (12), via the trimethylstannane precursor. Radiolabeling with either N-chlorosuccinimide or chloramine-T, in either MeCN or MeOH proceeded with a radiochemical conversion (RCC) of >80%. Subsequent deprotection only proved successful, among the tested conditions, when the radiolabeled Tz was stirred in 6-M HCl(aq.) at 60°C for 2.5 h. To the best of our knowledge, this is the first H-tetrazine labeled with iodine. In vivo investigations on the pretargeting ability of 12 are currently under way.


Assuntos
Compostos Heterocíclicos , Compostos Radiofarmacêuticos , Radioisótopos do Iodo , Química Click/métodos , Linhagem Celular Tumoral
7.
Chembiochem ; 23(23): e202200539, 2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36333105

RESUMO

Tetrazine (Tz)-trans-cyclooctene (TCO) ligation is an ultra-fast and highly selective reaction and it is particularly suited to label biomolecules under physiological conditions. As such, a 3 H-Tz based synthon would have wide applications for in vitro/ex vivo assays. In this study, we developed a 3 H-labeled Tz and characterized its potential for application to pretargeted autoradiography. Several strategies were explored to synthesize such a Tz. However, classical approaches such as reductive halogenation failed. For this reason, we designed a Tz containing an aldehyde and explored the possibility of reducing this group with NaBT4 . This approach was successful and resulted in [3 H]-(4-(6-(pyridin-2-yl)-1,2,4,5-tetrazin-3-yl)phenyl)methan-t-ol with a radiochemical yield of 22 %, a radiochemical purity of 96 % and a molar activity of 0.437 GBq/µmol (11.8 Ci/mmol). The compound was successfully applied to pretargeted autoradiography. Thus, we report the synthesis of the first 3 H-labeled Tz and its successful application as a labeling building block.


Assuntos
Compostos Heterocíclicos , Compostos Radiofarmacêuticos , Linhagem Celular Tumoral , Compostos Radiofarmacêuticos/química , Ciclo-Octanos/química
8.
Eur J Nucl Med Mol Imaging ; 49(8): 2668-2681, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35091755

RESUMO

BACKGROUND: Tumor-derived exosomes (TEX) have shown great potential for drug delivery and tumor targeting. Here, we developed a novel multi-drug loaded exosomes nanoprobe for combined antitumor chemotherapy and photodynamic therapy, and monitoring the drug delivery capabilities with pre-targeting technique. METHODS: TEX of human colorectal cancer HCT116 was prepared, and Doxorubicin and the photodynamic therapy agent 5-aminolevulinic acid (ALA) were loaded and named as TEX@DOX@ALA. Tumor uptake was first examined using fluorescence imaging of the fluorescent dye Cy5 (TEX@DOX@ALA@Cy5). Visualization of exosome aggregation in tumor were realized by positron-emission tomography/computed tomography (PET/CT) with pre-targeting technique. Tumor-bearing mice were first injected with TEX@DOX@ALA labeled with azide (N3) (TEX@DOX@ALA@N3), and then 68Ga-(2,2'-((6-amino-1-(4,7-bis (carboxymethyl)-1,4,7-triazonan-1-yl) hexan-2-yl) azanediyl) diacetic acid-dibenzocyclooctyne (68Ga-L-NETA-DBCO) was injected after 24 h for PET/CT imaging via in vivo click chemistry. For the antitumor therapy with photodynamic and/or chemotherapy, seven groups of tumor-bearing mice with different therapy were monitored, and the tumor size, animal weight and the survival time were recorded. Furthermore, the samples of blood and interested tissues (heart, lung, liver, kidney, and spleen) were harvested for hematological analysis and H&E staining. RESULTS: The drug loading process did not influence the structure or the function of the HCT116 TEX membranes. In a fluorescence imaging experiment, higher fluorescence could be seen in tumor after TEX@DOX@ALA@Cy5 injected, and reached the highest signal at 24 h. From PET/CT images with subcutaneous and orthotopic colon tumor-bearing mice, clear radioactivity could be seen in tumors, which suggested the successes of TEX accumulation in tumors. TEX@DOX@ALA group with photodynamic therapy and chemotherapy had the best tumor inhibition effect compared with the other groups, with the longest survival time (36 days, 37.5%). No significant damage was found on histological observation and the blood biochemical analysis, which suggested the safety of the multi-drug loaded exosomes. CONCLUSIONS: We successfully engineered an exosome-based nanoprobe integrating PET imaging components and therapeutic drugs. This drug-loaded exosome system may effectively target tumors and enable synergistic chemotherapeutic and photodynamic antitumor effects.


Assuntos
Exossomos , Neoplasias , Animais , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Radioisótopos de Gálio , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada
9.
Chemistry ; 28(61): e202201847, 2022 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-35851967

RESUMO

Combining nanotechnology and bioorthogonal chemistry for theranostic strategies offers the possibility to develop next generation nanomedicines. These materials are thought to increase therapeutic outcome and improve current cancer management. Due to their size, nanomedicines target tumors passively. Thus, they can be used for drug delivery purposes. Bioorthogonal chemistry allows for a pretargeting approach. Higher target-to-background drug accumulation ratios can be achieved. Pretargeting can also be used to induce internalization processes or trigger controlled drug release. Colloidal gold nanoparticles (AuNPs) have attracted widespread interest as drug delivery vectors within the last decades. Here, we demonstrate for the first time the possibility to successfully ligate AuNPs in vivo to pretargeted monoclonal antibodies. We believe that this possibility will facilitate the development of AuNPs for clinical use and ultimately, improve state-of-the-art patient care.


Assuntos
Ouro , Nanopartículas Metálicas , Humanos , Coloide de Ouro , Química Click , Linhagem Celular Tumoral , Anticorpos Monoclonais
10.
Mol Pharm ; 19(7): 2268-2278, 2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35700402

RESUMO

Pretargeting is a technique that uses macromolecules as targeting agents for nuclear imaging and therapy with the goal of reducing the radiation toxicity to healthy tissues often associated with directly radiolabeled macromolecules. In pretargeting, a macromolecule is radiolabeled in vivo at the target site using a radiolabeled small molecule (radioligand) that interacts with the macromolecule with high specificity. We report an investigation of host-guest chemistry-driven pretargeting using copper-64 radiolabeled ferrocene (Fc; guest) compounds and a cucurbit[7]uril (CB7; host) molecule functionalized carcinoembryonic antigen targeting hT84.66-M5A monoclonal antibody (CB7-M5A). Two novel ferrocene-based radioligands ([64Cu]Cu-NOTA-PEG3-Fc and [64Cu]Cu-NOTA-PEG7-Fc) were prepared, and their in vitro stability, pharmacokinetic in vivo profile in healthy mice, and pretargeting performance in a subcutaneous BxPC3 human pancreatic cancer cell xenograft mouse model were compared. The antibody dosing was optimized using a zirconium-89 radiolabeled M5A antibody ([89Zr]Zr-DFO-M5A) in a BxPC3 xenograft model, and the dosimetry of [89Zr]Zr-DFO-M5A and the pretargeting approach were compared. Finally, the effects of varying lag times up to 9 days between CB7-M5A and radioligand injection were investigated. In vivo pretargeting studies with both ferrocene radioligands resulted in specific tumor uptake (p = 0.0006 and p = 0.003) and also showed that the host-guest-based pretargeting approach excels with extended lag times up to 9 days with good tumor localization, suggesting that host-guest pretargeting may be suitable for use without clearing agents which have complicated clinical application of this technique. To our knowledge, the reported lag time of 9 days is the longest investigated lag time in any reported pretargeting studies.


Assuntos
Radioisótopos de Cobre , Imunoconjugados , Animais , Anticorpos Monoclonais/química , Linhagem Celular Tumoral , Radioisótopos de Cobre/química , Humanos , Imunoconjugados/farmacocinética , Metalocenos , Camundongos , Tomografia por Emissão de Pósitrons/métodos
11.
Mol Pharm ; 19(9): 3153-3162, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35635337

RESUMO

The last decade has witnessed the creation of a highly effective approach to in vivo pretargeting based on the inverse electron demand Diels-Alder (IEDDA) click ligation between tetrazine (Tz) and trans-cyclooctene (TCO). Despite the steady progression of this technology toward the clinic, concerns have persisted regarding whether this in vivo chemistry will work in humans given their larger size and blood volume. In this work, we describe the use of a 64Cu-labeled Tz radioligand ([64Cu]Cu-SarAr-Tz) and a TCO-bearing bisphosphonate (TCO-BP) for the pretargeted positron emission tomography (PET) imaging of osteodestructive lesions in a large animal model: companion dogs. First, in a small animal pilot study, healthy mice were injected with TCO-BP followed after 1 or 6 h by [64Cu]Cu-SarAr-Tz. PET images were collected 1, 6, and 24 h after the administration of [64Cu]Cu-SarAr-Tz, revealing that this approach produced high activity concentrations in the bone (>20 and >15%ID/g in the femur and humerus, respectively, at 24 h post injection) as well as high target-to-background contrast. Subsequently, companion dogs (n = 5) presenting with osteodestructive lesions were administered TCO-BP (5 or 10 mg/kg) followed 1 h later by [64Cu]Cu-SarAr-Tz (2.2-7.3 mCi; 81.4-270.1 MBq). PET scans were collected for each dog 4 h after the administration of the radioligand, and SUV values for the osteodestructive lesions, healthy bones, and kidneys were determined. In these animals, pretargeted PET clearly delineated healthy bone and produced very high activity concentrations in osteodestructive lesions. Low levels of uptake were observed in all healthy organs except for the kidneys and bladder due to the renal excretion of excess radioligand. Ultimately, this work not only illustrates that pretargeted PET with TCO-BP and [64Cu]Cu-SarAr-Tz is an effective tool for the visualization of osteodestructive lesions but also demonstrates for the first time that in vivo pretargeting based on IEDDA click chemistry is feasible in large animals.


Assuntos
Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Animais , Linhagem Celular Tumoral , Química Click , Ciclo-Octanos , Cães , Humanos , Camundongos , Projetos Piloto , Tomografia por Emissão de Pósitrons/métodos
12.
Chembiochem ; 22(1): 100-113, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-32935888

RESUMO

The advent of bioorthogonal chemistry has led to the development of powerful chemical tools that enable increasingly ambitious applications. In particular, these tools have made it possible to achieve what is considered to be the holy grail of many researchers involved in chemical biology: to perform unnatural chemical reactions within living organisms. In this minireview, we present an update of bioorthogonal reactions that have been carried out in animals for various applications. We outline the advances made in the understanding of fundamental biological processes, and the development of innovative imaging and therapeutic strategies using bioorthogonal chemistry.


Assuntos
Compostos Orgânicos/metabolismo , Animais , Química Click , Estrutura Molecular , Compostos Orgânicos/química
13.
J Labelled Comp Radiopharm ; 64(11): 428-439, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34330148

RESUMO

Amplification pretargeting has the potential to increase the tracer's accumulation in the tumor. This study aimed to develop a three-step amplification pretargeting strategy in nuclear medicine with a polymer conjugated with multiple copies of peptide nuclear acid (PNA). In this study, the tracer 18 F-labeled complementary PNA (18 F-cPNA) was prepared by click-chemistry with high radiochemical purity (>99%) and great stability in vitro. The PAMMA dendrimer generation 4 (G4) was conjugated with multiple copies of PNAs. The average number of PNA groups in the G4-PNA conjugate was determined by matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) and the accessibility to the 18 F-cPNA was identified by size-exclusion high-performance liquid chromatography (SE-HPLC). There were approximately 11.7 of 64 carboxyl groups modified with PNAs, of which more than 99% were accessible to 18 F-cPNA. 18 F-cPNA was added to a mixture of CC49-cPNA and G4-PNA, and the complex exhibited a single peak on high-performance liquid chromatography (HPLC) as evidence of complete hybridization between 18 F-cPNA and CC49-cPNA/G4-PNA. The LS174T tumor cells were incubated with CC49-cPNA followed by G4-PNA as an amplification platform before 18 F-cPNA was added to hybridize with CC49-cPNA/G4-PNA. Compared with conventional pretargeting without G4-PNA, the radioactivity signal was amplified about four times, which demonstrated that the dendrimer-PNA conjugate plays a crucial role in signal amplification.


Assuntos
Dendrímeros
14.
Molecules ; 26(15)2021 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-34361793

RESUMO

The pretargeting strategy has recently emerged in order to overcome the limitations of direct targeting, mainly in the field of radioimmunotherapy (RIT). This strategy is directly dependent on chemical reactions, namely bioorthogonal reactions, which have been developed for their ability to occur under physiological conditions. The Staudinger ligation, the copper catalyzed azide-alkyne cycloaddition (CuAAC) and the strain-promoted [3 + 2] azide-alkyne cycloaddition (SPAAC) were the first bioorthogonal reactions introduced in the literature. However, due to their incomplete biocompatibility and slow kinetics, the inverse-electron demand Diels-Alder (IEDDA) reaction was advanced in 2008 by Blackman et al. as an optimal bioorthogonal reaction. The IEDDA is the fastest bioorthogonal reaction known so far. Its biocompatibility and ideal kinetics are very appealing for pretargeting applications. The use of a trans-cyclooctene (TCO) and a tetrazine (Tz) in the reaction encouraged researchers to study them deeply. It was found that both reagents are sensitive to acidic or basic conditions. Furthermore, TCO is photosensitive and can be isomerized to its cis-conformation via a radical catalyzed reaction. Unfortunately, the cis-conformer is significantly less reactive toward tetrazine than the trans-conformation. Therefore, extensive research has been carried out to optimize both click reagents and to employ the IEDDA bioorthogonal reaction in biomedical applications.


Assuntos
Antineoplásicos/química , Química Click/métodos , Terapia de Alvo Molecular/métodos , Neoplasias/terapia , Radiossensibilizantes/química , Radioimunoterapia/métodos , Alcinos/química , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Azidas/química , Reação de Cicloadição/métodos , Ciclo-Octanos/química , Elétrons , Compostos Heterocíclicos com 1 Anel/química , Humanos , Concentração de Íons de Hidrogênio , Imunoconjugados/química , Imunoconjugados/farmacocinética , Imunoconjugados/farmacologia , Neoplasias/química , Neoplasias/imunologia , Neoplasias/patologia , Fotoquimioterapia/métodos , Radiossensibilizantes/farmacocinética , Radiossensibilizantes/farmacologia
15.
Angew Chem Int Ed Engl ; 60(33): 18082-18093, 2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34010512

RESUMO

Pretargeted imaging has emerged as a promising approach to advance nuclear imaging of malignant tumors. Herein, we combine the enzyme-mediated fluorogenic reaction and in situ self-assembly with the inverse electron demand Diels-Alder (IEDDA) reaction to develop an activatable pretargeted strategy for multimodality imaging. The trans-cyclooctene (TCO) bearing small-molecule probe, P-FFGd-TCO, can be activated by alkaline phosphatase and in situ self-assembles into nanoaggregates (FMNPs-TCO) retained on the membranes, permitting to (1) amplify near-infrared (NIR) fluorescence (FL) and magnetic resonance imaging (MRI) signals, and (2) enrich TCOs to promote IEDDA ligation. The Gallium-68 (68 Ga) labeled tetrazine can readily conjugate the tumor-retained FMNPs-TCO to enhance radioactivity uptake in tumors. Strong NIR FL, MRI, and positron emission tomography (PET) signals are concomitantly achieved, allowing for pretargeted multimodality imaging of ALP activity in HeLa tumor-bearing mice.


Assuntos
Fosfatase Alcalina/metabolismo , Ciclo-Octanos/metabolismo , Radioisótopos de Gálio/metabolismo , Imagem Multimodal , Bibliotecas de Moléculas Pequenas/metabolismo , Fosfatase Alcalina/química , Animais , Ciclo-Octanos/química , Radioisótopos de Gálio/química , Células HeLa , Humanos , Camundongos , Estrutura Molecular , Nanopartículas/química , Neoplasias Experimentais/diagnóstico por imagem , Tamanho da Partícula , Tomografia por Emissão de Pósitrons , Bibliotecas de Moléculas Pequenas/química
16.
Int J Mol Sci ; 21(4)2020 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-32098299

RESUMO

Pretargeting is widely explored in immunoPET as a strategy to reduce radiation exposure of non-target organs and allow the use of short-lived radionuclides that would not otherwise be compatible with the slow pharmacokinetic profiles of antibodies. Here we investigate a pretargeting strategy based on gallium-68 and the chelator THPMe as a high-affinity pair capable of combining in vivo. After confirming the ability of THPMe to bind 68Ga in vivo at low concentrations, the bifunctional THPMe-NCS was conjugated to a humanised huA33 antibody targeting the A33 glycoprotein. Imaging experiments performed in nude mice bearing A33-positive SW1222 colorectal cancer xenografts compared pretargeting (100 µg of THPMe-NCS-huA33, followed after 24 h by 8-10 MBq of 68Ga3+) with both a directly labelled radioimmunoconjugate (89Zr-DFO-NCS-huA33, 88 µg, 7 MBq) and a 68Ga-only negative control (8-10 MBq of 68Ga3+). Imaging was performed 25 h after antibody administration (1 h after 68Ga3+ administration for negative control). No difference between pretargeting and the negative control was observed, suggesting that pretargeting via metal chelation is not feasible using this model. However, significant accumulation of "unchelated" 68Ga3+ in the tumour was found (12.9 %ID/g) even without prior administration of THPMe-NCS-huA33, though tumour-to-background contrast was impaired by residual activity in the blood. Therefore, the 68Ga-only experiment was repeated using THPMe (20 µg, 1 h after 68Ga3+ administration) to clear circulating 68Ga3+, producing a three-fold improvement of the tumour-to-blood activity concentration ratio. Although preliminary, these results highlight the potential of THPMe as a 68Ga clearing agent in imaging applications with gallium citrate.


Assuntos
Anticorpos/metabolismo , Quelantes/farmacocinética , Imunoconjugados/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Animais , Anticorpos/química , Linhagem Celular Tumoral , Quelantes/química , Feminino , Radioisótopos de Gálio/química , Radioisótopos de Gálio/metabolismo , Radioisótopos de Gálio/farmacocinética , Xenoenxertos , Humanos , Imunoconjugados/química , Imunoconjugados/metabolismo , Taxa de Depuração Metabólica , Camundongos Endogâmicos BALB C , Camundongos Nus , Estrutura Molecular , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/metabolismo , Distribuição Tecidual
17.
Mol Pharm ; 16(5): 2259-2263, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30912951

RESUMO

Pretargeted radioimmunotherapy (PRIT) based on the inverse electron demand Diels-Alder reaction has shown promise in murine models of disease, yet the radiation dosimetry of this approach must be optimized to make it a viable clinical option. To this end, we have leveraged two recent developments in pretargeted imaging-dendritic scaffolds and masking agents-to improve the dosimetric profile of a proof-of-concept PRIT system that is based on the huA33 antibody, a 177Lu-labeled tetrazine radioligand ([177Lu]Lu-DOTA-PEG7-Tz), and a mouse model of A33 antigen-expressing colorectal carcinoma. Pretargeting using an huA33 immunoconjugate bearing a trans-cyclooctene-decorated dendritic scaffold (sshuA33-DEN-TCO) produced significantly higher tumoral activity concentrations at 120 h post-injection (23.0 ± 2.2 %ID/g) than those achieved with an analogous, dendrimer-lacking immunoconjugate (12.7 ± 2.6 %ID/g). However, pretargeting using sshuA33-DEN-TCO also resulted in increased activity concentrations in the blood at the same time point (1.9 ± 0.4 %ID/g) compared to the dendrimer-lacking construct (0.7 ± 0.2 %ID/g), thereby curtailing improvements to the tumor-to-blood therapeutic ratio of the system. In order to circumvent this issue, a tetrazine-labeled, dextran-based masking agent (Tz-DP) was injected prior to the radioligand to prevent the ligation between [177Lu]Lu-DOTA-PEG7-Tz and circulating immunoconjugate. This approach dramatically decreased the absorbed dose to the blood but also attenuated the absorbed dose to the tumor and increased the absorbed dose to the lungs. Ultimately, these data suggest that dendritic scaffolds and masking agents could be used to improve the dosimetry of PRIT, but the combination of these technologies will require extensive optimization.


Assuntos
Neoplasias Colorretais/terapia , Radioimunoterapia/métodos , Animais , Anticorpos/imunologia , Linhagem Celular Tumoral , Estudos de Coortes , Neoplasias Colorretais/patologia , Meios de Contraste/química , Reação de Cicloadição/métodos , Ciclo-Octanos/química , Dendrímeros/química , Modelos Animais de Doenças , Feminino , Compostos Heterocíclicos com 1 Anel/química , Humanos , Imunoconjugados/uso terapêutico , Lutécio/química , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Nus , Radioisótopos/química , Radiometria/métodos , Compostos Radiofarmacêuticos/uso terapêutico , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Mol Pharm ; 16(10): 4416-4421, 2019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31483993

RESUMO

Recent years have played witness to the advent of nuclear theranostics: the synergistic use of "matched pair" radiopharmaceuticals for diagnostic imaging and targeted radiotherapy. In this investigation, we report the extension of this concept to in vivo pretargeting based on the rapid and bioorthogonal inverse electron demand Diels-Alder reaction between tetrazine (Tz) and trans-cyclooctene (TCO). We demonstrate that a single injection of a TCO-modified immunoconjugate can be used as a platform for pretargeted PET imaging and radiotherapy via the sequential administration of a pair of Tz-bearing radioligands labeled with the positron-emitting radiometal copper-64 (t1/2 ≈ 12.7 h) and the beta-emitting radiometal lutetium-177 (t1/2 ≈ 6.7 days). More specifically, a mouse model of human colorectal carcinoma received a dose of the A33 antigen-targeting immunoconjugate huA33-TCO, followed 24 and 48 h later by injections of [64Cu]Cu-SarAr-Tz and [177Lu]Lu-DOTA-PEG7-Tz, respectively. This approach produces high activity concentrations of both radioligands in tumor tissue (16.4 ± 2.7 %ID/g for [64Cu]Cu-SarAr-Tz at 48 h post-injection and 18.1 ± 2.1 %ID/g for [177Lu]Lu-DOTA-PEG7-Tz at 120 h post-injection) as well as promising tumor-to-healthy organ activity concentration ratios. Ultimately, we believe that this work could not only have important implications in nuclear theranostics-most excitingly with isotopologue-based radioligand pairs such as [64Cu]Cu-SarAr-Tz and [67Cu]Cu-SarAr-Tz-but also in the delivery of fractionated doses during pretargeted radioimmunotherapy.


Assuntos
Neoplasias Colorretais/diagnóstico por imagem , Neoplasias Colorretais/terapia , Imunoconjugados/metabolismo , Glicoproteínas de Membrana/imunologia , Radioimunoterapia/métodos , Compostos Radiofarmacêuticos/metabolismo , Nanomedicina Teranóstica , Animais , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Radioisótopos de Cobre/química , Ciclo-Octanos/química , Feminino , Compostos Heterocíclicos com 1 Anel/química , Humanos , Imunoconjugados/química , Lutécio/química , Lutécio/metabolismo , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons , Radioisótopos/química , Radioisótopos/metabolismo , Compostos Radiofarmacêuticos/química , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Bioorg Med Chem Lett ; 29(8): 986-990, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30795854

RESUMO

Pretargeted nuclear imaging based on the ligation between tetrazines and nano-sized targeting agents functionalized with trans-cyclooctene (TCO) has recently been shown to improve both imaging contrast and dosimetry in nuclear imaging of nanomedicines. Herein, we describe the improved radiosynthesis of a 11C-labeled tetrazine ([11C]AE-1) and its preliminary evaluation in both mice and pigs. Pretargeted imaging in mice was carried out using both a new TCO-functionalized polyglutamic acid and a previously reported TCO-functionalized bisphosphonate system as targeting agents. Unfortunately, pretargeted imaging was not successful using these targeting agents in pair with [11C]AE-1. However, brain imaging in pig indicated that the tracer crossed the blood-brain-barrier. Hence, we suggest that this tetrazine scaffold could be used as a starting point for the development of pretargeted brain imaging, which has so far been a challenging task.


Assuntos
Radioisótopos de Carbono/química , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , Tetrazóis/química , Animais , Encéfalo/diagnóstico por imagem , Radioisótopos de Carbono/metabolismo , Difosfonatos/química , Marcação por Isótopo , Camundongos , Neoplasias/diagnóstico por imagem , Ácido Poliglutâmico/química , Compostos Radiofarmacêuticos/metabolismo , Suínos , Tetrazóis/metabolismo , Distribuição Tecidual
20.
Bioorg Med Chem Lett ; 29(14): 1791-1798, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31101473

RESUMO

During the last four decades, nuclear medicine has undergone enormous growth, and positron emission tomography (PET) has been in the driving seat for most of the time. 18F-fluorodeoxyglucose (18F-FDG) is the most widely used agent for the detection of hibernating myocardium and metabolically active cancer tissue. But its cost and limited availability are the main limitations. For a long time different researchers and groups of pharmacists have tried to label glucose with a cheaper and long-acting radionuclide like 99mTc. However, they failed to achieve this goal owing to the chemical complexity of 99mTc and the lack of maintaining the physiological activity of diagnostic compounds. A pre-targeting strategy based on strain-promoted [3 + 2] azide-alkyne cycloaddition (SPAAC) reaction was applied to solve this problem. Functional click synthons were synthesized: 2-azido-2-deoxy-d-glucose (GlucN3) as a glucose analogue, and N- (2- (2- (2- (bis (pyridin-2-ylmethyl) amino) ethoxy) ethoxy) ethyl-2- (6H-11,12-didehydrodibenzo [a,e] cycloocten-5-ylideneaminooxy) acetamide (C7) as a 99mTc(CO)3 labeling and azido-binding group. The results of biodistribution experiments in mice bearing S180 tumor show the relatively high tumor/blood ratio (up to 2.95) and tumor/muscle ratio (up to 6.37), and both of them decreases significantly in the glucose blocking experiment. It indicates that GlucN3 behaves similarly to glucose and that in vivo SPAAC reactions can occur effectively. It is supposed that this pre-targeting strategy can indeed enhance target specificity and may be used for glucose metabolism imaging in tumor diagnosis.


Assuntos
Glucose/metabolismo , Compostos de Organotecnécio/química , Animais , Camundongos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa