Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.807
Filtrar
Más filtros

Colección Oncologia Uruguay
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 630(8015): 206-213, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38778111

RESUMEN

Targeted radionuclide therapy, in which radiopharmaceuticals deliver potent radionuclides to tumours for localized irradiation, has addressed unmet clinical needs and improved outcomes for patients with cancer1-4. A therapeutic radiopharmaceutical must achieve both sustainable tumour targeting and fast clearance from healthy tissue, which remains a major challenge5,6. A targeted ligation strategy that selectively fixes the radiopharmaceutical to the target protein in the tumour would be an ideal solution. Here we installed a sulfur (VI) fluoride exchange (SuFEx) chemistry-based linker on radiopharmaceuticals to prevent excessively fast tumour clearance. When the engineered radiopharmaceutical binds to the tumour-specific protein, the system undergoes a binding-to-ligation transition and readily conjugates to the tyrosine residues through the 'click' SuFEx reaction. The application of this strategy to a fibroblast activation protein (FAP) inhibitor (FAPI) triggered more than 80% covalent binding to the protein and almost no dissociation for six days. In mice, SuFEx-engineered FAPI showed 257% greater tumour uptake than did the original FAPI, and increased tumour retention by 13-fold. The uptake in healthy tissues was rapidly cleared. In a pilot imaging study, this strategy identified more tumour lesions in patients with cancer than did other methods. SuFEx-engineered FAPI also successfully achieved targeted ß- and α-radionuclide therapy, causing nearly complete tumour regression in mice. Another SuFEx-engineered radioligand that targets prostate-specific membrane antigen (PSMA) also showed enhanced therapeutic efficacy. Considering the broad scope of proteins that can potentially be ligated to SuFEx warheads, it might be possible to adapt this strategy to other cancer targets.


Asunto(s)
Terapia Molecular Dirigida , Neoplasias de la Próstata , Radioisótopos , Radiofármacos , Animales , Humanos , Masculino , Ratones , Antígenos de Superficie/química , Antígenos de Superficie/metabolismo , Línea Celular Tumoral , Fluoruros/química , Fluoruros/metabolismo , Glutamato Carboxipeptidasa II/química , Glutamato Carboxipeptidasa II/metabolismo , Ligandos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/química , Terapia Molecular Dirigida/métodos , Proyectos Piloto , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/radioterapia , Radioisótopos/uso terapéutico , Radiofármacos/química , Radiofármacos/uso terapéutico , Radiofármacos/metabolismo , Radiofármacos/farmacocinética , Compuestos de Azufre/química , Compuestos de Azufre/metabolismo , Tirosina/metabolismo , Tirosina/química , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Nature ; 606(7912): 102-108, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35344982

RESUMEN

The advent of total-body positron emission tomography (PET) has vastly broadened the range of research and clinical applications of this powerful molecular imaging technology1. Such possibilities have accelerated progress in fluorine-18 (18F) radiochemistry with numerous methods available to 18F-label (hetero)arenes and alkanes2. However, access to 18F-difluoromethylated molecules in high molar activity is mostly an unsolved problem, despite the indispensability of the difluoromethyl group for pharmaceutical drug discovery3. Here we report a general solution by introducing carbene chemistry to the field of nuclear imaging with a [18F]difluorocarbene reagent capable of a myriad of 18F-difluoromethylation processes. In contrast to the tens of known difluorocarbene reagents, this 18F-reagent is carefully designed for facile accessibility, high molar activity and versatility. The issue of molar activity is solved using an assay examining the likelihood of isotopic dilution on variation of the electronics of the difluorocarbene precursor. Versatility is demonstrated with multiple [18F]difluorocarbene-based reactions including O-H, S-H and N-H insertions, and cross-couplings that harness the reactivity of ubiquitous functional groups such as (thio)phenols, N-heteroarenes and aryl boronic acids that are easy to install. The impact is illustrated with the labelling of highly complex and functionalized biologically relevant molecules and radiotracers.


Asunto(s)
Radioisótopos de Flúor , Hidrocarburos Fluorados , Tomografía de Emisión de Positrones , Radiofármacos , Ácidos Borónicos/química , Radioisótopos de Flúor/química , Hidrocarburos Fluorados/química , Imagen Molecular , Tomografía de Emisión de Positrones/métodos , Radiofármacos/química
3.
EMBO J ; 40(22): e107757, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34636430

RESUMEN

Positron emission tomography (PET) allows biomolecular tracking but PET monitoring of brain networks has been hampered by a lack of suitable reporters. Here, we take advantage of bacterial dihydrofolate reductase, ecDHFR, and its unique antagonist, TMP, to facilitate in vivo imaging in the brain. Peripheral administration of radiofluorinated and fluorescent TMP analogs enabled PET and intravital microscopy, respectively, of neuronal ecDHFR expression in mice. This technique can be used to the visualize neuronal circuit activity elicited by chemogenetic manipulation in the mouse hippocampus. Notably, ecDHFR-PET allows mapping of neuronal projections in non-human primate brains, demonstrating the applicability of ecDHFR-based tracking technologies for network monitoring. Finally, we demonstrate the utility of TMP analogs for PET studies of turnover and self-assembly of proteins tagged with ecDHFR mutants. These results establish opportunities for a broad spectrum of previously unattainable PET analyses of mammalian brain circuits at the molecular level.


Asunto(s)
Encéfalo/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Radiofármacos/química , Tetrahidrofolato Deshidrogenasa/genética , Animales , Encéfalo/citología , Callithrix , Radioisótopos de Carbono/química , Radioisótopos de Flúor/química , Genes Reporteros , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Imagen Molecular/métodos , Red Nerviosa/diagnóstico por imagen , Proteínas/análisis , Proteínas/metabolismo , Radiofármacos/síntesis química , Tetrahidrofolato Deshidrogenasa/metabolismo , Trimetoprim/análogos & derivados , Trimetoprim/química
4.
Acc Chem Res ; 57(6): 933-944, 2024 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-38501206

RESUMEN

Nuclear medicine harnesses radioisotopes for the diagnosis and treatment of disease. While the isotopes 99mTc and 111In have enabled the clinical diagnosis of millions of patients over the past 3 decades, more recent clinical translation of numerous 68Ga/177Lu-based radiopharmaceuticals for diagnostic imaging and therapy underscores the clinical utility of metal-based radiopharmaceuticals in mainstream cancer treatment. In addition to such established radionuclides, advancements in radioisotope production have enabled the production of radionuclides with a broad range of half-lives and emission properties of interest for nuclear medicine. Chemical means to form kinetically inert, in vivo-compatible species that can be modified with disease-targeting vectors is imperative. This presents a challenge for radiosiotopes of elements where the aqueous chemistry is still underdeveloped and poorly understood. Here, we discuss our efforts to date in exploring the aqueous, radioactive coordination chemistry of highly Lewis acidic metal ions and how our discoveries apply to the diagnosis and treatment of cancer in preclinical models of disease. The scope of this Account includes approaches to aqueous coordination of to-date understudied highly Lewis acidic metal ions with radioisotopes of emerging interest and the modulation of well-understood coordination environments of radio-coordination complexes to induce metal-catalyzed reactivity for separation and pro-drug applications.First, we discuss the development of seven-coordinate, small-cavity macrocyclic chelator platform mpatcn/picaga as an exemplary case study, which forms robust complexes with 44Sc/47Sc isotopes. Due to the high chemical hardness and pronounced Lewis acidity of the Sc3+ ion, the displacement of ternary ligand H2O by 18/natF- can be achieved to form an inert Sc-18/natF bond. Corresponding coordination complex natSc-18F is in vivo compatible and forms a theranostic tetrad with corresponding 44Sc/47Sc, 177Lu complexes all exhibiting homologous biodistribution profiles. Another exceptionally hard, highly Lewis acidic ion with underdeveloped aqueous chemistry and emerging interest in nuclear medicine is 45Ti4+. To develop de novo approaches to the mononuclear chelation of this ion under aqueous conditions, we employed a fragment-based bidentate ligand screening approach which identified two leads. The screen successfully predicted the formation of [45Ti][Ti(TREN-CAM)], a Ti-triscatechol complex that exhibits remarkable in vivo stability. Furthermore, the fragment-based screen also identified approaches that enabled solid-phase separation of Ti4+ and Sc3+ of interest in streamlining the isotope production of 45Ti and accessing new ways to separate 44Ti/44Sc for the development of a long-lived generator system. In addition to establishing the inert chelation of Ti4+ and Sc3+, we introduce controlled, metal-induced reactivity of corresponding coordination complexes on macroscopic and radiotracer scales. Metal-mediated autolytic amide bond cleavage (MMAAC) enables the temperature-dependent release of high-molar-activity, ready-to-inject radiopharmaceuticals; cleavage is selectively triggered by coordinated trivalent Lewis acid nat/68Ga3+ or Sc3+. Following the scope of reactivity and mechanistic studies, we validated MMAAC for the synthesis of high-molar-activity radiopharmaceuticals to image molecular targets with low expression and metal-mediated prodrug hydrolysis in vivo.This Account summarizes how developing the aqueous coordination chemistry and tuning the chemical reactivity of metal ions with high Lewis acidity at the macroscopic and tracer scales directly apply to the radiopharmaceutical synthesis with clinical potential.


Asunto(s)
Complejos de Coordinación , Medicina Nuclear , Humanos , Radiofármacos/química , Ácidos de Lewis , Complejos de Coordinación/química , Ligandos , Distribución Tisular , Radioisótopos/química , Quelantes/química , Metales , Iones
5.
Acc Chem Res ; 57(9): 1421-1433, 2024 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-38666539

RESUMEN

Molecular imaging with antibodies radiolabeled with positron-emitting radionuclides combines the affinity and selectivity of antibodies with the sensitivity of Positron Emission Tomography (PET). PET imaging allows the visualization and quantification of the biodistribution of the injected radiolabeled antibody, which can be used to characterize specific biological interactions in individual patients. This characterization can provide information about the engagement of the antibody with a molecular target such as receptors present in elevated levels in tumors as well as providing insight into the distribution and clearance of the antibody. Potential applications of clinical PET with radiolabeled antibodies include identifying patients for targeted therapies, characterization of heterogeneous disease, and monitoring treatment response.Antibodies often take several days to clear from the blood pool and localize in tumors, so PET imaging with radiolabeled antibodies requires the use of a radionuclide with a similar radioactive half-life. Zirconium-89 is a positron-emitting radionuclide that has a radioactive half-life of 78 h and relatively low positron emission energy that is well suited to radiolabeling antibodies. It is essential that the zirconium-89 radionuclide be attached to the antibody through chemistry that provides an agent that is stable in vivo with respect to the dissociation of the radionuclide without compromising the biological activity of the antibody.This Account focuses on our research using a simple derivative of the bacterial siderophore desferrioxamine (DFO) with a squaramide ester functional group, DFO-squaramide (DFOSq), to link the chelator to antibodies. In our work, we produce conjugates with an average ∼4 chelators per antibody, and this does not compromise the binding of the antibody to the target. The resulting antibody conjugates of DFOSq are stable and can be easily radiolabeled with zirconium-89 in high radiochemical yields and purity. Automated methods for the radiolabeling of DFOSq-antibody conjugates have been developed to support multicenter clinical trials. Evaluation of several DFOSq conjugates with antibodies and low molecular weight targeting agents in tumor mouse models gave PET images with high tumor uptake and low background. The promising preclinical results supported the translation of this chemistry to human clinical trials using two different radiolabeled antibodies. The potential clinical impact of these ongoing clinical trials is discussed.The use of DFOSq to radiolabel relatively low molecular weight targeting molecules, peptides, and peptide mimetics is also presented. Low molecular weight molecules typically clear the blood pool and accumulate in target tissue more rapidly than antibodies, so they are usually radiolabeled with positron-emitting radionuclides with shorter radioactive half-lives such as fluorine-18 (t1/2 ∼ 110 min) or gallium-68 (t1/2 ∼ 68 min). Radiolabeling peptides and peptide mimetics with zirconium-89, with its longer radioactive half-life (t1/2 = 78 h), could facilitate the centralized manufacture and distribution of radiolabeled tracers. In addition, the ability to image patients at later time points with zirconium-89 based agents (e.g. 4-24 h after injection) may also allow the delineation of small or low-uptake disease sites as the delayed imaging results in increased clearance of the tracer from nontarget tissue and lower background signal.


Asunto(s)
Deferoxamina , Tomografía de Emisión de Positrones , Quinina/análogos & derivados , Radioisótopos , Circonio , Circonio/química , Radioisótopos/química , Deferoxamina/química , Tomografía de Emisión de Positrones/métodos , Animales , Humanos , Ratones , Radiofármacos/química , Neoplasias/diagnóstico por imagen
6.
J Am Chem Soc ; 146(25): 17517-17529, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38869959

RESUMEN

Despite the widespread use of hydrophilic building blocks to incorporate 18F and improve tracer pharmacokinetics, achieving effective leaving group-mediated nucleophilic 18F-fluorination in water (excluding 18F/19F-exchange) remains a formidable challenge. Here, we present a water-compatible SN2 leaving group-mediated 18F-fluorination method employing preconjugated "AquaF" (phosphonamidic fluorides) building blocks. Among 19 compact tetracoordinated pentavalent P(V)-F candidates, the "AquaF" building blocks exhibit superior water solubility, sufficient capacity for 18F-fluorination in water, and excellent in vivo metabolic properties. Two nitropyridinol leaving groups, identified from a pool of leaving group candidates that further enhance the precursor water solubility, enable 18F-fluorination in water with a 10-2 M-1 s-1 level reaction rate constant (surpassing the 18F/19F-exchange) at room temperature. With the exergonic concerted SN2 18F-fluorination mechanism confirmed, this 18F-fluorination method achieves ∼90% radiochemical conversions and reaches a molar activity of 175 ± 40 GBq/µmol (using 12.2 GBq initial activity) in saline for 12 "AquaF"-modified proof-of-concept functional substrates and small-molecule 18F-tracers. [18F]AquaF-Flurpiridaz demonstrates significantly improved radiochemical yield and molar activity compared to 18F-Flurpiridaz, alongside enhanced cardiac uptake and heart/liver ratio in targeted myocardial perfusion imaging, providing a comprehensive illustration of "AquaF" building blocks-assisted water-compatible SN2 18F-fluorination of small-molecule radiotracers.


Asunto(s)
Radioisótopos de Flúor , Halogenación , Agua , Radioisótopos de Flúor/química , Agua/química , Animales , Radiofármacos/química , Radiofármacos/síntesis química , Ratones , Tomografía de Emisión de Positrones , Solubilidad , Estructura Molecular , Trazadores Radiactivos
7.
Anal Chem ; 96(29): 11725-11733, 2024 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-38975941

RESUMEN

AIMS: PET myocardial perfusion imaging (MPI) is the gold standard for the noninvasive diagnosis of ischemic myocardial. Construction of 18F-labeled PET MPI probe showed benefits to reduce the imaging cost, and enhance the image quality and patient-friendliness. METHODS: Two 18F-labeled MPI probes (18F-BoMPI) were developed. Detailed in vitro/vivo evaluation including photophysical properties, in vitro stability, myocardial cell uptake kinetics and mechanisms, cytotoxicity and IC50, biodistribution and plasma clearance curve were investigated. Resting and stressing myocardial perfusion PET imaging were performed in healthy and myocardial ischemic mice. RESULTS: 18F-BoMPI could be quickly labeled and easily postprocessed, and demonstrated excellent in vitro stability. Cell assays indicated that 18F-BoMPI exhibited mitochondria-targeting but potential-independent myocardial uptake. In vivo evaluation revealed the effective myocardial uptake and rapid background clearance. PET MPI confirmed effective probe accumulation in the healthy heart, but rapidly clearance in the background, making heart clearly delineated in the images. Ischemic myocardial could be clearly distinguished as the region of radioactivity sparsity in PET MPI. CONCLUSION: The 18F-labeled probes showed great potentials to reduce the practicability threshold of PET MPI.


Asunto(s)
Radioisótopos de Flúor , Imagen de Perfusión Miocárdica , Tomografía de Emisión de Positrones , Radiofármacos , Animales , Radioisótopos de Flúor/química , Imagen de Perfusión Miocárdica/métodos , Ratones , Radiofármacos/química , Distribución Tisular , Humanos , Masculino , Isquemia Miocárdica/diagnóstico por imagen
8.
Chembiochem ; 25(11): e202400152, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38695673

RESUMEN

Positron emission tomography imaging of misfolded proteins with high-affinity and selective radioligands has played a vital role in expanding our knowledge of neurodegenerative diseases such as Parkinson's and Alzheimer's disease. The pathogenesis of Huntington's disease, a CAG trinucleotide repeat disorder, is similarly linked to the presence of protein fibrils formed from mutant huntingtin (mHTT) protein. Development of mHTT fibril-specific radioligands has been limited by the lack of structural knowledge around mHTT and a dearth of available hit compounds for medicinal chemistry refinement. Over the past decade, the CHDI Foundation, a non-for-profit scientific management organisation has orchestrated a large-scale screen of small molecules to identify high affinity ligands of mHTT, with lead compounds now reaching clinical maturity. Here we describe the mHTT radioligands developed to date and opportunities for further improvement of this radiotracer class.


Asunto(s)
Proteína Huntingtina , Tomografía de Emisión de Positrones , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Proteína Huntingtina/química , Ligandos , Humanos , Agregado de Proteínas/efectos de los fármacos , Mutación , Enfermedad de Huntington/diagnóstico por imagen , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética , Radiofármacos/química
9.
Bioconjug Chem ; 35(2): 232-244, 2024 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-38215469

RESUMEN

Transition-metal-mediated bioconjugation chemistry has been used extensively to design and synthesize molecular probes to visualize, characterize, and quantify biological processes within intact living organisms at the cellular and subcellular levels. We demonstrate the development and validation of chemoselective [18F]fluoro-arylation chemistry of cysteine residues using Pd-mediated S-arylation chemistry with 4-[18F]fluoroiodobenzene ([18F]FIB) as an aryl electrophile. The novel bioconjugation technique proceeded in excellent radiochemical yields of 73-96% within 15 min under ambient and aqueous reaction mixture conditions, representing a versatile novel tool for decorating peptides and peptidomimetics with short-lived positron emitter 18F. The chemoselective S-arylation of several peptides and peptidomimetics containing multiple reactive functional groups confirmed the versatility and functional group compatibility. The synthesis and radiolabeling of a novel prostate-specific membrane antigen (PSMA) binding radioligand [18F]6 was accomplished using the novel labeling protocol. The validation of radioligand [18F]6 in a preclinical prostate cancer model with PET resulted in favorable accumulation and retention in PSMA-expressing LNCaP tumors. At the same time, a significantly lower salivary gland uptake was observed compared to clinical PSMA radioligand [18F]PSMA-1007. This finding coincides with ongoing discussions about the molecular basis of the off-target accumulation of PSMA radioligands currently used for clinical imaging and therapy of prostate cancer.


Asunto(s)
Peptidomiméticos , Neoplasias de la Próstata , Masculino , Humanos , Paladio , Cisteína , Línea Celular Tumoral , Neoplasias de la Próstata/patología , Glutamato Carboxipeptidasa II/metabolismo , Antígenos de Superficie , Péptidos , Radiofármacos/química , Tomografía de Emisión de Positrones/métodos
10.
Bioconjug Chem ; 35(9): 1335-1342, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39172920

RESUMEN

Single-domain antibodies, or nanobodies (Nbs), are promising biomolecules for use in molecular imaging due to their excellent affinity, specificity, and fast clearance from the blood. Given their short blood half-life, pairing Nbs with short-lived imaging radioisotopes is desirable. Because fluorine-18 (18F) is routinely used for clinical imaging, it is an attractive radioisotope for Nbs. We report a novel sortase-based, site-specific 18F-labeling method applied to three nanobodies. Labeled nanobodies were synthesized either by a two-step indirect radiolabeling method in one pot or by a one-step direct labeling method using a sortase-mediated conjugation of either the radiolabeled chelator (H-GGGK((±)-Al[18F]FH3RESCA)-NH2) or the unlabeled chelator (H-GGGK((±)-H3RESCA)-NH2) followed by labeling with Al[18F]F, respectively. The overall radiochemical yields were 15-43% (n = 22, decay-corrected) in 70 min (indirect labeling) and 23-58% (n = 12, decay-corrected) in 50 min (direct labeling). The radiochemical purities of the labeled nanobodies prepared by both methods were >98% with a specific activity of 400-600 Ci/mmol (n = 22) for each of the three Nbs tested and exhibited excellent stability profiles under physiological conditions. This simple, site-specific, reproducible, and generalizable 18F-labeling method to prepare nanobodies (Nb-Al[18F]F-RESCA) or other low molecular weight biomolecules can easily be adopted in various settings for preclinical and clinical studies.


Asunto(s)
Aminoaciltransferasas , Radioisótopos de Flúor , Anticuerpos de Dominio Único , Radioisótopos de Flúor/química , Anticuerpos de Dominio Único/química , Aminoaciltransferasas/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas Bacterianas/química , Marcaje Isotópico/métodos , Quelantes/química , Humanos , Radiofármacos/química
11.
Bioconjug Chem ; 35(9): 1402-1416, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39185789

RESUMEN

The Al18F-labeling approach offers a one-step access to radiofluorinated biomolecules by mimicking the labeling process for radiometals. Although these labeling conditions are considered to be mild compared to classic radiofluorinations, improvements of the chelating units have led to the discovery of (±)-H3RESCA, which allows Al18F-labeling already at ambient temperature. While the suitability of (±)-H3RESCA for functionalization and radiofluorination of proteins is well established, its use for small molecules or peptides is less explored. Herein, we advanced this acyclic pentadentate ligand by introducing an alkyne moiety for the late-stage functionalization of biomolecules via click chemistry. We show that in addition to Al18F-labeling, the cyclohexanediamine triazole (CHDT) moiety allows stable complexation of 68Ga and 111In. Three novel CHDT-functionalized PSMA inhibitors were synthesized and their Al18F-, 68Ga-, and 111In-labeled analogs were subjected to a detailed in vitro radiopharmacological characterization. Stability studies in vitro in human serum revealed among others a high kinetic inertness of all radiometal complexes. Furthermore, the Al18F-labeled PSMA ligands were characterized for their biodistribution in a LNCaP derived tumor xenograft mouse model by PET imaging. One radioligand, Al[18F]F-CHDT-PSMA-1, bearing a small azidoacetyl linker at the glutamate-urea-lysine motif, provided an in vivo performance comparable to that of [18F]PSMA-1007 but with even higher tumor-to-blood and tumor-to-muscle ratios at 120 min p.i. Overall, our results highlight the suitability of the novel CHDT moiety for functionalization and radiolabeling of small molecules or peptides with Al18F, 68Ga, and 111In and the triazole ring seems to entail favorable pharmacokinetic properties for molecular imaging purposes.


Asunto(s)
Radioisótopos de Flúor , Triazoles , Animales , Triazoles/química , Triazoles/farmacocinética , Humanos , Ratones , Radioisótopos de Flúor/química , Radioisótopos de Galio/química , Radiofármacos/química , Radiofármacos/farmacocinética , Radiofármacos/síntesis química , Masculino , Línea Celular Tumoral , Química Clic , Distribución Tisular
12.
Bioconjug Chem ; 35(9): 1309-1317, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-38954733

RESUMEN

Fibroblast activation protein (FAP) has recently gained significant attention as a promising tumor biomarker for both diagnosis and therapeutic applications. A series of radiopharmaceuticals based on fibroblast activation protein inhibitors (FAPIs) have been developed and translated into the clinic. Though some of them such as radiolabeled FAPI-04 probes have achieved favorable in vivo imaging performance, further improvement is still highly desired for obtaining radiopharmaceuticals with a high theranostics potential. In this study, we innovatively designed an FAPI ligand SMIC-3002 by changing the core quinoline motif of FAPI-04 to the quinolinium scaffold. The engineered molecule was further radiolabeled with 68Ga to generate a positron emission tomography (PET) probe, [68Ga]Ga-SMIC-3002, which was then evaluated in vitro and in vivo. [68Ga]Ga-SMIC-3002 demonstrated high in vitro stability, nanomolar affinity for FAP (8 nM for protein, 23 nM for U87MG cells), and specific uptake in FAP-expressing tumors, with a tumor/muscle ratio of 19.1 and a tumor uptake of 1.48 ± 0.03 ID/g% at 0.5 h in U87MG tumor-bearing mice. In summary, the quinolinium scaffold can be successfully used for the development of the FAP-targeted tracer. [68Ga]Ga-SMIC-3002 not only shows high potential for clinical translation but also offers insights into designing a new generation of FAPI tracers.


Asunto(s)
Radioisótopos de Galio , Tomografía de Emisión de Positrones , Tomografía de Emisión de Positrones/métodos , Humanos , Animales , Ratones , Radioisótopos de Galio/química , Línea Celular Tumoral , Radiofármacos/química , Compuestos de Quinolinio/química , Proteínas de la Membrana/metabolismo , Endopeptidasas/metabolismo , Serina Endopeptidasas/metabolismo , Distribución Tisular , Ratones Desnudos
13.
Bioconjug Chem ; 35(8): 1160-1165, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39023912

RESUMEN

Photoredox is a powerful synthetic tool in organic chemistry and has been widely used in various fields, including nuclear medicine and molecular imaging. In particular, acridinium-based organophotoredox radiolabeling has significantly impacted the production and discovery of positron emission tomography (PET) agents. Despite their extensive use in preclinical research, no PET agents synthesized by acridinium photoredox labeling have been tested in humans. [18F]FDOPA is clinically used for tumor diagnosis and the evaluation of neuropsychiatric disorders, but its application is limited by complex synthesis methods, the need for expensive modules, and/or the high cost of consumable materials/cassettes. In this report, we integrated a photoredox labeling unit with an automated module and produced [18F]FDOPA for human study. This research not only represents the first human study of a PET agent generated by acridinium-based organophotoredox reactions but also demonstrates the safety of this novel labeling method, serving as a milestone/reference for the clinical translation of other PET agents generated by this technique in the future.


Asunto(s)
Dihidroxifenilalanina , Oxidación-Reducción , Tomografía de Emisión de Positrones , Humanos , Tomografía de Emisión de Positrones/métodos , Dihidroxifenilalanina/análogos & derivados , Dihidroxifenilalanina/química , Radiofármacos/química , Radiofármacos/síntesis química , Acridinas/química , Procesos Fotoquímicos , Radioisótopos de Flúor/química
14.
Bioconjug Chem ; 35(5): 665-673, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38598424

RESUMEN

Enhancing the accumulation and retention of small-molecule probes in tumors is an important way to achieve accurate cancer diagnosis and therapy. Enzyme-stimulated macrocyclization of small molecules possesses great potential for enhanced positron emission tomography (PET) imaging of tumors. Herein, we reported an 18F-labeled radiotracer [18F]AlF-RSM for legumain detection in vivo. The tracer was prepared by a one-step aluminum-fluoride-restrained complexing agent ([18F]AlF-RESCA) method with high radiochemical yield (RCY) (88.35 ± 3.93%) and radiochemical purity (RCP) (>95%). More notably, the tracer can be transformed into a hydrophobic macrocyclic molecule under the joint action of legumain and reductant. Simultaneously, the tracer could target legumain-positive tumors and enhance accumulation and retention in tumors, resulting in the amplification of PET imaging signals. The enhancement of radioactivity enables PET imaging of legumain activity with high specificity. We envision that, by combining this highly efficient 18F-labeled strategy with our intramolecular macrocyclization reaction, a range of radiofluorinated tracers can be designed for tumor PET imaging and early cancer diagnosis in the future.


Asunto(s)
Cisteína Endopeptidasas , Radioisótopos de Flúor , Tomografía de Emisión de Positrones , Tomografía de Emisión de Positrones/métodos , Radioisótopos de Flúor/química , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/análisis , Animales , Ciclización , Ratones , Humanos , Radiofármacos/química , Línea Celular Tumoral , Ratones Endogámicos BALB C , Fluoruros/química , Ratones Desnudos
15.
Bioconjug Chem ; 35(5): 567-574, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38634516

RESUMEN

The exploration of pharmaceutically active agents and positron emission tomography (PET) tracers targeting CXCR4 has been a focal point in cancer research given its pivotal role in the development and progression of various cancers. While significant strides have been made in PET imaging with radiometal-labeled tracers, the landscape of 18F-labeled small molecule tracers remains relatively limited. Herein, we introduce a novel and promising derivative, [18F]SFB-AMD3465, as a targeted PET tracer for CXCR4. The compound was synthesized by modifying the pyridine ring of AMD3465, which was subsequently labeled with 18F using [18F]SFB. The study provides comprehensive insights into the design, synthesis, and biological evaluation of [18F]SFB-AMD3465. In vitro and in vivo assessments demonstrated the CXCR4-dependent, specific, and sensitive uptake of [18F]SFB-AMD3465 in the CXCR4-overexpressing 4T1 cell line and the corresponding xenograft-bearing mouse model. These findings contribute to bridging the gap in 18F-labeled PET tracers for CXCR4 and underscore the potential of [18F]SFB-AMD3465 as a PET radiotracer for in vivo CXCR4 imaging.


Asunto(s)
Radioisótopos de Flúor , Tomografía de Emisión de Positrones , Receptores CXCR4 , Animales , Receptores CXCR4/análisis , Receptores CXCR4/metabolismo , Tomografía de Emisión de Positrones/métodos , Ratones , Radioisótopos de Flúor/química , Femenino , Línea Celular Tumoral , Humanos , Piridinas/química , Piridinas/farmacocinética , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Radiofármacos/química , Radiofármacos/farmacocinética , Distribución Tisular
16.
J Biol Inorg Chem ; 29(2): 187-199, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38607392

RESUMEN

Aß42 plaque formation is one of the preliminary pathologic events that occur post traumatic brain injury (TBI) which is also among the most noteworthy hallmarks of AD. Their pre symptomatic detection is therefore vital for better disease management. Chalcone-picolinic acid chelator derivative, 6-({[(6-carboxypyridin-2-yl)methyl](2-{4-[(2E)-3-[4-(dimethyl amino)phenyl]prop-2-enoyl]phenoxy}ethyl)amino}methyl)pyridine-2-carboxylic acid, Py-chal was synthesized to selectively identify amyloid plaques formed post head trauma using SPECT imaging by stable complexation to 99mTc with > 97% efficiency without compromising amyloid specificity. The binding potential of the Py-chal ligand to amyloid plaques remained high as confirmed by in vitro binding assay and photophysical spectra. Further, the Py-chal complex stained amyloid aggregates in the brain sections of rmTBI mice model. In vivo scintigraphy in TBI mice model displayed high uptake followed by high retention while the healthy rabbits displayed higher brain uptake followed by a rapid washout attributed to absence of amyloid plaques. Higher uptake in brain of TBI model was also confirmed by ex vivo biodistribution analysis wherein brain uptake of 3.38 ± 0.2% ID/g at 2 min p.i. was observed for TBI mice model. This was followed by prolonged retention and more than twofold higher activity as compared to sham mice brain. This preliminary data suggests the specificity of the radiotracer for amyloid detection post head trauma and applicability of 99mTc labeled Py-chal complex for TBI-induced ß-amyloid SPECT imaging.


Asunto(s)
Péptidos beta-Amiloides , Lesiones Traumáticas del Encéfalo , Chalcona , Tomografía Computarizada de Emisión de Fotón Único , Animales , Masculino , Ratones , Péptidos beta-Amiloides/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Lesiones Traumáticas del Encéfalo/metabolismo , Chalcona/química , Chalcona/farmacología , Traumatismos Craneocerebrales/diagnóstico por imagen , Compuestos de Organotecnecio/química , Compuestos de Organotecnecio/farmacocinética , Radiofármacos/química , Radiofármacos/farmacocinética , Radiofármacos/síntesis química , Tecnecio/química , Tecnecio/farmacología , Distribución Tisular
17.
Eur J Nucl Med Mol Imaging ; 51(11): 3322-3333, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38771516

RESUMEN

PURPOSE: Accumulating evidence suggests that neurotensin (NTS) and neurotensin receptors (NTSRs) play key roles in lung cancer progression by triggering multiple oncogenic signaling pathways. This study aims to develop Cu-labeled neurotensin receptor 1 (NTSR1)-targeting agents with the potential for both imaging and therapeutic applications. METHOD: A series of neurotensin receptor antagonists (NRAs) with variable propylamine (PA) linker length and different chelators were synthesized, including [64Cu]Cu-CB-TE2A-iPA-NRA ([64Cu]Cu-4a-c, i = 1, 2, 3), [64Cu]Cu-NOTA-2PA-NRA ([64Cu]Cu-4d), [64Cu]Cu-DOTA-2PA-NRA ([64Cu]Cu-4e, also known as [64Cu]Cu-3BP-227), and [64Cu]Cu-DOTA-VS-2PA-NRA ([64Cu]Cu-4f). The series of small animal PET/CT were conducted in H1299 lung cancer model. The expression profile of NTSR1 was also confirmed by IHC using patient tissue samples. RESULTS: For most of the compounds studied, PET/CT showed prominent tumor uptake and high tumor-to-background contrast, but the tumor retention was strongly influenced by the chelators used. For previously reported 4e, [64Cu]Cu-labeled derivative showed initial high tumor uptake accompanied by rapid tumor washout at 24 h. The newly developed [64Cu]Cu-4d and [64Cu]Cu-4f demonstrated good tumor uptake and tumor-to-background contrast at early time points, but were less promising in tumor retention. In contrast, our lead compound [64Cu]Cu-4b demonstrated 9.57 ± 1.35, 9.44 ± 2.38 and 9.72 ± 4.89%ID/g tumor uptake at 4, 24, and 48 h p.i., respectively. Moderate liver uptake (11.97 ± 3.85, 9.80 ± 3.63, and 7.72 ± 4.68%ID/g at 4, 24, and 48 h p.i.) was observed with low uptake in most other organs. The PA linker was found to have a significant effect on drug distribution. Compared to [64Cu]Cu-4b, [64Cu]Cu-4a had a lower background, including a greatly reduced liver uptake, while the tumor uptake was only moderately reduced. Meanwhile, [64Cu]Cu-4c showed increased uptake in both the tumor and the liver. The clinical relevance of NTSR1 was also demonstrated by the elevated tumor expression in patient tissue samples. CONCLUSIONS: Through the side-by-side comparison, [64Cu]Cu-4b was identified as the lead agent for further evaluation based on its high and sustained tumor uptake and moderate liver uptake. It can not only be used to efficiently detect NTSR1 expression in lung cancer (for diagnosis, patient screening, and treatment monitoring), but also has the great potential to treat NTSR-positive lesions once chelating to the beta emitter 67Cu.


Asunto(s)
Quelantes , Radioisótopos de Cobre , Radiofármacos , Receptores de Neurotensina , Animales , Receptores de Neurotensina/metabolismo , Ratones , Quelantes/química , Radiofármacos/farmacocinética , Radiofármacos/química , Distribución Tisular , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones , Marcaje Isotópico
18.
Eur J Nucl Med Mol Imaging ; 51(11): 3334-3345, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38787395

RESUMEN

PURPOSE: The advancement of heterodimeric tracers, renowned for their high sensitivity, marks a significant trend in the development of radiotracers for cancer diagnosis. Our prior work on [68Ga]Ga-HX01, a heterodimeric tracer targeting CD13 and integrin αvß3, led to its approval for phase I clinical trials by the China National Medical Production Administration (NMPA). However, its fast clearance and limited tumor retention pose challenges for broader clinical application in cancer treatment. This study aims to develop a new radiopharmaceutical with increased tumor uptake and prolonged retention, rendering it a potential therapeutic candidate. METHODS: New albumin binder-conjugated compounds were synthesized based on the structure of HX01. In vitro and in vivo evaluation of these new compounds were performed after labelling with 68Ga. Small-animal PET/CT imaging were conducted at different time points at 0.5-6 h post injection (p.i.) using BxPC-3 xenograft mice models. The one with the best imaging performance was further radiolabeled with 177Lu for small-animal SPECT/CT and ex vivo biodistribution investigation. RESULTS: We have synthesized novel albumin binder-conjugated compounds, building upon the structure of HX01. When radiolabeled with 68Ga, all compounds demonstrated improved pharmacokinetics (PK). Small-animal PET/CT studies revealed that these new albumin binder-conjugated compounds, particularly [68Ga]Ga-L6, exhibited significantly enhanced tumor accumulation and retention compared with [68Ga]Ga-L0 without an albumin binder. [68Ga]Ga-L6 outperformed [68Ga]Ga-L7, a compound developed using a previously reported albumin binder. Furthermore, [177Lu]Lu-L6 demonstrated rapid clearance from normal tissues, high tumor uptake, and prolonged retention in small-animal SPECT/CT and biodistribution studies, positioning it as an ideal candidate for radiotherapeutic applications. CONCLUSION: A new integrin αvß3 and CD13 targeting compound was screened out. This compound bears a novel albumin binder and exhibits increased tumor uptake and prolonged tumor retention in BxPC-3 tumors and low background in normal organs, making it a perfect candidate for radiotherapy when radiolabeled with 177Lu.


Asunto(s)
Integrina alfaVbeta3 , Radiofármacos , Animales , Radiofármacos/farmacocinética , Radiofármacos/química , Radiofármacos/uso terapéutico , Integrina alfaVbeta3/metabolismo , Ratones , Humanos , Distribución Tisular , Línea Celular Tumoral , Albúminas/química , Tomografía Computarizada por Tomografía de Emisión de Positrones , Femenino , Neoplasias/diagnóstico por imagen , Neoplasias/radioterapia
19.
Eur J Nucl Med Mol Imaging ; 51(9): 2761-2773, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38561515

RESUMEN

Fibroblast activation protein (FAP) has emerged as a highly promising target for cancer diagnostic imaging and targeted radionuclide therapy. To exploit the therapeutic potential of suitably radiolabeled FAP inhibitors (FAPIs), this study presents the design and synthesis of a series of FAPI dimers to increase tumor uptake and retention. Preclinical evaluation and a pilot clinical PET imaging study were conducted to screen the lead compound with the potential for radionuclide therapy. METHODS: Three new FAPI dimers were synthesized by linking two quinoline-based FAPIs with different spacers. The in vitro binding affinity and preclinical small animal PET imaging of the compounds were compared with their monomeric counterparts, FAPI-04 and FAPI-46. The lead compound, [68Ga]Ga -LNC1013, was then evaluated in a pilot clinical PET imaging study involving seven patients with gastrointestinal cancer. RESULTS: The three newly synthesized FAPI homodimers had high binding affinity and specificity in vitro and in vivo. Small animal PET imaging and biodistribution studies showed that [68Ga]Ga-LNC1013 had persistent tumor retention for at least 4 h, also higher uptake than the other two dimers and the monomer counterparts, making it the lead compound to enter clinical investigation. In the pilot clinical PET imaging study, seven patients were enrolled. The effective dose of [68Ga]Ga-LNC1013 was 8.24E-03 mSv/MBq. The human biodistribution of [68Ga]Ga-LNC1013 demonstrated prominent tumor uptake and good tumor-to-background contrast. [68Ga]Ga-LNC1013 PET imaging showed potential in capturing primary and metastatic lesions and outperforming 18F-FDG PET in detecting pancreatic and esophageal cancers. The SUVmax for lesions with [68Ga]Ga-FAPI-46 decreased over time, whereas [68Ga]Ga-LNC1013 exhibited persistently high tumor uptake from 1 to 4 h post-injection. CONCLUSION: Dimerization is an effective strategy to produce FAPI derivatives with favorable tumor uptake, long tumor retention, and imaging contrast over its monomeric counterpart. We demonstrated that [68Ga]Ga-LNC1013, the lead compound without any piperazine moiety, had superior diagnostic potential over [68Ga]Ga-FAPI-46 and 18F-FDG, suggesting the future potential of LNC1013 for radioligand therapy of FAP-positive cancers.


Asunto(s)
Radioisótopos de Galio , Humanos , Animales , Ratones , Radioisótopos de Galio/química , Línea Celular Tumoral , Femenino , Distribución Tisular , Tomografía de Emisión de Positrones/métodos , Masculino , Dimerización , Radiofármacos/farmacocinética , Radiofármacos/química , Persona de Mediana Edad , Investigación Biomédica Traslacional , Anciano , Proteínas de la Membrana , Endopeptidasas , Quinolinas
20.
Eur J Nucl Med Mol Imaging ; 51(9): 2663-2671, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38570359

RESUMEN

PURPOSE: A probe for targeted alpha therapy (TAT) using the RGD peptide (Ga-DOTA-K([211At]APBA)-c(RGDfK) ([211At]1)) with albumin-binding moiety (ABM) was recently developed. [211At]1 highly accumulated in tumors and significantly inhibited tumor growth in U-87 MG tumor-bearing mice. However, high [211At]1 retention in blood may cause critical adverse events, such as hematotoxicity. Therefore, we attempted to accelerate the blood clearance of [211At]1 by competitively inhibiting the binding of [211At]1 to albumin to modulate the pharmacokinetics of the former. METHODS: To evaluate the effects of albumin-binding inhibitors in normal mice, sodium 4-(4-iodophenyl)butanoate at 2, 5, or 10 molar equivalents of blood albumin was administered at 1-h postinjection of [211At]1. The biodistribution of [211At]1, SPECT/CT imaging of [67Ga]Ga-DOTA-K(IPBA)-c(RGDfK) ([67Ga]2), and the therapeutic effects of [211At]1 were compared with or without IPBA administration in U-87 MG tumor-bearing mice. RESULTS: Blood radioactivity of [211At]1 was decreased in a dose-dependent manner with IPBA in normal mice. In U-87 MG tumor-bearing mice, the blood radioactivity and accumulation in nontarget tissues of [211At]1 were decreased by IPBA. Meanwhile, tumor [211At]1 accumulation was not changed at 3-h postinjection of IPBA. In SPECT/CT imaging of [67Ga]2, IPBA administration dramatically decreased radioactivity in nontarget tissues, and only tumor tissue was visualized. In therapeutic experiments, [211At]1 with IPBA injected-group significantly inhibited tumor growth compared to the control group. CONCLUSION: IPBA administration (as an albumin-binding inhibitor) could modulate the pharmacokinetics and enhance the therapeutic effects of [211At]1.


Asunto(s)
Oligopéptidos , Animales , Ratones , Oligopéptidos/farmacocinética , Oligopéptidos/química , Distribución Tisular , Línea Celular Tumoral , Humanos , Radiofármacos/farmacocinética , Radiofármacos/química , Albúminas/química , Albúminas/farmacocinética , Unión Proteica , Masculino , Marcaje Isotópico , Albúmina Sérica/química , Femenino , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA