Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 93.460
Filtrer
1.
J Nanobiotechnology ; 22(1): 419, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39014410

RÉSUMÉ

BACKGROUND: Iron oxide nanoparticles (IONPs) have been cleared by the Food and Drug Administration (FDA) for various clinical applications, such as tumor-targeted imaging, hyperthermia therapy, drug delivery, and live-cell tracking. However, the application of IONPs as T1 contrast agents has been restricted due to their high r2 values and r2/r1 ratios, which limit their effectiveness in T1 contrast enhancement. Notably, IONPs with diameters smaller than 5 nm, referred to as extremely small-sized IONPs (ESIONs), have demonstrated potential in overcoming these limitations. To advance the clinical application of ESIONs as T1 contrast agents, we have refined a scale-up process for micelle encapsulation aimed at improving the hydrophilization of ESIONs, and have carried out comprehensive in vivo biodistribution and preclinical toxicity assessments. RESULTS: The optimization of the scale-up micelle-encapsulation process, specifically employing Tween60 at a concentration of 10% v/v, resulted in ESIONs that were uniformly hydrophilized, with an average size of 9.35 nm and a high purification yield. Stability tests showed that these ESIONs maintained consistent size over extended storage periods and dispersed effectively in blood and serum-mimicking environments. Relaxivity measurements indicated an r1 value of 3.43 mM- 1s- 1 and a favorable r2/r1 ratio of 5.36, suggesting their potential as T1 contrast agents. Biodistribution studies revealed that the ESIONs had extended circulation times in the bloodstream and were primarily cleared via the hepatobiliary route, with negligible renal excretion. We monitored blood clearance and organ distribution using positron emission tomography and magnetic resonance imaging (MRI). Additionally, MRI signal variations in a dose-dependent manner highlighted different behaviors at varying ESIONs concentrations, implying that optimal dosages might be specific to the intended imaging application. Preclinical safety evaluations indicated that ESIONs were tolerable in rats at doses up to 25 mg/kg. CONCLUSIONS: This study effectively optimized a scale-up process for the micelle encapsulation of ESIONs, leading to the production of hydrophilic ESIONs at gram-scale levels. These optimized ESIONs showcased properties conducive to T1 contrast imaging, such as elevated r1 relaxivity and a reduced r2/r1 ratio. Biodistribution study underscored their prolonged bloodstream presence and efficient clearance through the liver and bile, without significant renal involvement. The preclinical toxicity tests affirmed the safety of the ESIONs, supporting their potential use as T1 contrast agent with versatile clinical application.


Sujet(s)
Produits de contraste , Nanoparticules magnétiques d'oxyde de fer , Imagerie par résonance magnétique , Micelles , Taille de particule , Animaux , Produits de contraste/composition chimique , Produits de contraste/pharmacocinétique , Distribution tissulaire , Imagerie par résonance magnétique/méthodes , Nanoparticules magnétiques d'oxyde de fer/composition chimique , Nanoparticules magnétiques d'oxyde de fer/toxicité , Souris , Rats , Mâle , Humains , Femelle
2.
J Extracell Vesicles ; 13(7): e12489, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39016198

RÉSUMÉ

Recently, extracellular vesicles (EVs) have been developed as therapeutic targets for various diseases. Biodistribution is crucial for EVs intended for therapeutic purposes because it can determine the degree of on- and off-target effects. This study aimed to explore techniques to evaluate the biodistribution of unmodified EVs. We devised a novel quantitative polymerase chain reaction (qPCR)-based assay to detect unmodified EVs by targeting mitochondrial deoxyribonucleic acid (mtDNA), a constituent of EVs. We focused on specific mtDNA regions that exhibited homologous variations distinct from their rodent mtDNA counterparts to establish this analytical approach. Herein, we successfully designed primers and probes targeting human and rodent mtDNA sequences and developed a highly specific and sensitive qPCR method. Furthermore, the quantification range of EVs isolated from various cells differed based on the manufacturer and cell source. IRDye 800CW-labelled Expi293F EV mimetics were administered to the animals via the tail vein to compare the imaging test and mtDNA-qPCR results. The results obtained from imaging tests and mtDNA-qPCR to investigate EV biodistribution patterns revealed differences. The results revealed that our newly developed method effectively determined the biodistribution of unmodified EVs with high sensitivity and reproducibility.


Sujet(s)
ADN mitochondrial , Vésicules extracellulaires , Vésicules extracellulaires/métabolisme , Animaux , ADN mitochondrial/métabolisme , Humains , Distribution tissulaire , Souris , Rats , Mitochondries/métabolisme
3.
ACS Appl Mater Interfaces ; 16(28): 35925-35935, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38950334

RÉSUMÉ

The development of efficient theranostic nanoagents for the precise diagnosis and targeted therapy of glioblastoma (GBM) remains a big challenge. Herein, we designed and developed porphyrin-based organic nanoparticles (PNP NPs) with strong emission in the near-infrared IIa window (NIR-IIa) for orthotopic GBM theranostics. PNP NPs possess favorable photoacoustic and photothermal properties, high photostability, and low toxicity. After modification with the RGD peptide, the obtained PNPD NPs exhibited enhanced blood-brain barrier (BBB) penetration capability and GBM targeting ability. NIR-IIa imaging was employed to monitor the in vivo biodistribution and accumulation of the nanoparticles, revealing a significant enhancement in penetration depth and signal-to-noise ratio. Both in vitro and in vivo results demonstrated that PNPD NPs effectively inhibited the proliferation of tumor cells and induced negligible side effects in normal brain tissues. In general, the work presented a kind of brain-targeted porphyrin-based NPs with NIR-IIa fluorescence for orthotopic glioblastoma theranostics, showing promising prospects for clinical translation.


Sujet(s)
Glioblastome , Nanoparticules , Porphyrines , Nanomédecine théranostique , Glioblastome/imagerie diagnostique , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Animaux , Nanoparticules/composition chimique , Humains , Porphyrines/composition chimique , Porphyrines/pharmacologie , Souris , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Rayons infrarouges , Distribution tissulaire , Barrière hémato-encéphalique/métabolisme , Souris nude , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Fluorescence
4.
J Pharm Biomed Anal ; 248: 116326, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38959756

RÉSUMÉ

Antibiotic-associated diarrhea (AAD) is a common side effect of antibiotic therapy, characterized by intestinal inflammation which reduces the quality of life of patients. Xianglian Pill (XLP) has long been used to treat abdominal pain, diarrhea, bacillary dysentery and enteritis. Studies found that XLP has curative effect on AAD; however, the chemical constituents and mechanism of XLP have not been fully elucidated because of the lack of in vitro and in vivo studies. In this study, ultra-high performance liquid chromatography mass spectrometry method (UPLC-Q-Exactive-Orbitrap-HRMS) was used to examine the components of the XLP. Then, the binding between active compounds and the key targets was studied using network pharmacology and molecular docking. A comparative tissue distribution study was established for the simultaneous determination of the 10 active components in healthy and AAD mouse models. Forty-six components were characterized from XLP. According to the network pharmacology degree value, a prediction was made that encompassed 42 components and 14 core targets, which were intricately involved in crucial biological pathways, such as the AGE-RAGE signaling, cellular senescence, and MAPK signaling. Tissue distribution analysis showed that the 10 components were widely distributed in the heart, liver, spleen, lungs, kidneys, small intestine, and large intestine of mice, with varying concentrations in healthy and AAD mice. Molecular docking analysis also indicated that the active compounds in the tissue distribution could bind tightly to key targets of network pharmacological studies. This study provides a reference for further investigations of the relationships between the chemical components and pharmacological activities of XLP.


Sujet(s)
Antibactériens , Diarrhée , Modèles animaux de maladie humaine , Médicaments issus de plantes chinoises , Simulation de docking moléculaire , Animaux , Souris , Diarrhée/induit chimiquement , Diarrhée/traitement médicamenteux , Mâle , Chromatographie en phase liquide à haute performance/méthodes , Médicaments issus de plantes chinoises/composition chimique , Médicaments issus de plantes chinoises/pharmacologie , Distribution tissulaire , Pharmacologie des réseaux/méthodes
5.
J Pharm Biomed Anal ; 248: 116288, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38981330

RÉSUMÉ

Germacrone and curdione are germacrane-type sesquiterpenoids that are widely distributed and have extensive pharmacological activities; they are the main constituents of 'Xing-Nao-Jing Injection' (XNJ). Studies on the metabolic features of germacrane-type sesquiterpenoids are limited. In this study, the metabolites of germacrone and curdione were characterized by UHPLC-Q-Exactive Oribitrap mass spectrometry after they were orally administered to rats. In total, 60 and 76 metabolites were found and preliminarily identified in rats administered germacrone and curdione, respectively, among which at least 123 potential new compounds were included. New metabolic reactions of germacrane-type sesquiterpenoids were identified, which included oxidation (+4 O and +5 O), ethylation, methyl-sulfinylation, vitamin C conjugation, and cysteine conjugation reactions. Among the 136 metabolites (including 113 oxidation metabolites, two glucuronidation, two methylation, nine methyl-sulfinylation, three ethylation, six cysteine conjugation, and one Vitamin C conjugation metabolites), 32 metabolites were detected in nine organs, and the stomach, intestine, liver, kidneys, and small intestine were the main organs for the distribution of these metabolites. All 136 metabolites were detected in urine and 64 of them were found in feces. The results of this study not only contribute to research on in vivo processes related to germacrane-type sesquiterpenoids but also provide a strong foundation for a better understanding of in vivo processes and the effective forms of germacrone, curdione, and XNJ.


Sujet(s)
Médicaments issus de plantes chinoises , Rat Sprague-Dawley , Sesquiterpènes de type germacrane , Animaux , Sesquiterpènes de type germacrane/métabolisme , Rats , Médicaments issus de plantes chinoises/pharmacocinétique , Médicaments issus de plantes chinoises/métabolisme , Médicaments issus de plantes chinoises/administration et posologie , Mâle , Chromatographie en phase liquide à haute performance/méthodes , Distribution tissulaire , Administration par voie orale , Fèces/composition chimique
6.
Sci Rep ; 14(1): 15960, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987294

RÉSUMÉ

Non-invasive imaging of GSK-3 expression in the brain will help to understand the role of GSK-3 in disease pathology and progression. Herein, we report the radiosynthesis and evaluation of two novel isonicotinamide based 18F labeled PET probes, [18F]2 and [18F]6 for noninvasive imaging of GSK3. Among the developed PET probes, the in vitro blood-brain permeability coefficient of 2 (38 ± 20 × 10-6 cm/s, n = 3) was found to be better than 6 (8.75 ± 3.90 × 10-6 cm/s, n = 5). The reference compounds 2 and 6 showed nanomolar affinity towards GSK-3α and GSK-3ß. PET probe [18F]2 showed higher stability (100%) in mouse and human serums compared to [18F]6 (67.01 ± 4.93%, n = 3) in mouse serum and 66.20 ± 6.38%, n = 3) in human serum at 120 min post incubation. The in vivo imaging and blocking studies were performed in wild-type mice only with [18F]2 due to its observed stability. [18F]2 showed a SUV of 0.92 ± 0.28 (n = 6) in mice brain as early as 5 min post-injection followed by gradual clearance over time.


Sujet(s)
Encéphale , Radio-isotopes du fluor , Glycogen Synthase Kinase 3 , Tomographie par émission de positons , Tomographie par émission de positons/méthodes , Animaux , Humains , Souris , Radio-isotopes du fluor/composition chimique , Encéphale/imagerie diagnostique , Encéphale/métabolisme , Glycogen Synthase Kinase 3/métabolisme , Radiopharmaceutiques/composition chimique , Radiopharmaceutiques/synthèse chimique , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/imagerie diagnostique , Distribution tissulaire
7.
J Nanobiotechnology ; 22(1): 394, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965594

RÉSUMÉ

DNA nanostructures have long been developed for biomedical purposes, but their controlled delivery in vivo proposes a major challenge for disease theranostics. We previously reported that DNA nanostructures on the scales of tens and hundreds nanometers showed preferential renal excretion or kidney retention, allowing for sensitive evaluation and effective protection of kidney function, in response to events such as unilateral ureter obstruction or acute kidney injury. Encouraged by the positive results, we redirected our focus to the liver, specifically targeting organs noticeably lacking DNA materials, to explore the interaction between DNA nanostructures and the liver. Through PET imaging, we identified SDF and M13 as DNA nanostructures exhibiting significant accumulation in the liver among numerous candidates. Initially, we investigated and assessed their biodistribution, toxicity, and immunogenicity in healthy mice, establishing the structure-function relationship of DNA nanostructures in the normal murine. Subsequently, we employed a mouse model of liver ischemia-reperfusion injury (IRI) to validate the nano-bio interactions of SDF and M13 under more challenging pathological conditions. M13 not only exacerbated hepatic oxidative injury but also elevated local apoptosis levels. In contrast, SDF demonstrated remarkable ability to scavenge oxidative responses in the liver, thereby mitigating hepatocyte injury. These compelling results underscore the potential of SDF as a promising therapeutic agent for liver-related conditions. This aimed to elucidate their roles and mechanisms in liver injury, providing a new perspective for the biomedical applications of DNA nanostructures.


Sujet(s)
ADN , Foie , Nanostructures , Lésion d'ischémie-reperfusion , Animaux , Lésion d'ischémie-reperfusion/traitement médicamenteux , Souris , Foie/métabolisme , ADN/composition chimique , Nanostructures/composition chimique , Mâle , Distribution tissulaire , Souris de lignée C57BL , Apoptose/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques
8.
J Med Chem ; 67(14): 11827-11840, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39013156

RÉSUMÉ

Fibroblast activation protein (FAP) has attracted considerable attention as a possible target for the radiotherapy of solid tumors. Unfortunately, initial efforts to treat solid tumors with FAP-targeted radionuclides have yielded only modest clinical responses, suggesting that further improvements in the molecular design of FAP-targeted radiopharmaceutical therapies (RPT) are warranted. In this study, we report several advances on the previously described FAP6 radioligand that increase tumor retention and accelerate healthy tissue clearance. Seven FAP6 derivatives with different linkers or albumin binders were synthesized, radiolabeled, and investigated for their effects on binding and cellular uptake. The radioligands were then characterized in 4T1 tumor-bearing Balb/c mice using both single-photon emission computed tomography (SPECT) and ex vivo biodistribution analyses to identify the conjugate with the best tumor retention and tumor-to-healthy organ ratios. The results reveal an optimized FAP6 radioligand that exhibits efficacy and safety properties that potentially justify its translation into the clinic.


Sujet(s)
Endopeptidases , Gelatinases , Protéines membranaires , Souris de lignée BALB C , Radiopharmaceutiques , Serine endopeptidases , Tomographie par émission monophotonique , Animaux , Endopeptidases/métabolisme , Souris , Distribution tissulaire , Protéines membranaires/métabolisme , Radiopharmaceutiques/composition chimique , Radiopharmaceutiques/synthèse chimique , Radiopharmaceutiques/pharmacocinétique , Radiopharmaceutiques/pharmacologie , Radiopharmaceutiques/usage thérapeutique , Gelatinases/métabolisme , Femelle , Serine endopeptidases/métabolisme , Lignée cellulaire tumorale , Humains , Ligands
9.
Drug Deliv ; 31(1): 2380538, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39044468

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic inflammatory joint disease accompanied by energy depletion and accumulation of reactive oxygen species (ROS). Inorganic nanoparticles (NPs) offer great promise for the treatment of RA because they mostly have functions beyond being drug carriers. However, conventional nanomaterials become coated with a protein corona (PC) or lose their cargo prematurely in vivo, reducing their therapeutic efficacy. To avoid these problems, we loaded methotrexate (MTX) into hollow structured manganese dioxide nanoparticles (H-MnO2 NPs), then coated them with a 'pseudo-corona' of human serum albumin (HSA) at physiological concentrations to obtain HSA-MnO2@MTX NPs. Efficacy of MTX, MnO2@MTX, and HSA-MnO2@MTX NPs was compared in vitro and in vivo. Compared to MnO2@MTX, HSA-coated NPs were taken up better by lipopolysaccharide-activated RAW264.7 and were more effective at lowering levels of pro-inflammatory cytokines and preventing ROS accumulation. HSA-MnO2@MTX NPs were also more efficient at blocking the proliferation and migration of fibroblast-like synoviocytes from rats with collagen-induced arthritis. In this rat model, HSA-MnO2@MTX NPs showed better biodistribution than other treatments, specifically targeting the ankle joint. Furthermore, HSA-MnO2@MTX NPs reduced swelling in the paw, regulated pro-inflammatory cytokine production, and limited cartilage degradation and signs of inflammation. These results establish the therapeutic potential of HSA-MnO2@MTX NPs against RA.


Sujet(s)
Polyarthrite rhumatoïde , Vecteurs de médicaments , Composés du manganèse , Méthotrexate , Nanoparticules , Oxydes , Espèces réactives de l'oxygène , Sérum-albumine humaine , Animaux , Composés du manganèse/composition chimique , Oxydes/composition chimique , Polyarthrite rhumatoïde/traitement médicamenteux , Souris , Rats , Nanoparticules/composition chimique , Sérum-albumine humaine/composition chimique , Humains , Méthotrexate/pharmacologie , Méthotrexate/administration et posologie , Méthotrexate/pharmacocinétique , Méthotrexate/composition chimique , Espèces réactives de l'oxygène/métabolisme , Cellules RAW 264.7 , Vecteurs de médicaments/composition chimique , Mâle , Arthrite expérimentale/traitement médicamenteux , Distribution tissulaire , Antirhumatismaux/pharmacologie , Antirhumatismaux/administration et posologie , Antirhumatismaux/pharmacocinétique , Rat Sprague-Dawley , Cytokines/métabolisme
10.
Pharm Res ; 41(7): 1391-1400, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38981900

RÉSUMÉ

PURPOSE: Evaluation of distribution kinetics is a neglected aspect of pharmacokinetics. This study examines the utility of the model-independent parameter whole body distribution clearance (CLD) in this respect. METHODS: Since mammillary compartmental models are widely used, CLD was calculated in terms of parameters of this model for 15 drugs. The underlying distribution processes were explored by assessment of relationships to pharmacokinetic parameters and covariates. RESULTS: The model-independence of the definition of the parameter CLD allowed a comparison of distributional properties of different drugs and provided physiological insight. Significant changes in CLD were observed as a result of drug-drug interactions, transporter polymorphisms and a diseased state. CONCLUSION: Total distribution clearance CLD is a useful parameter to evaluate distribution kinetics of drugs. Its estimation as an adjunct to the model-independent parameters clearance and steady-state volume of distribution is advocated.


Sujet(s)
Taux de clairance métabolique , Modèles biologiques , Pharmacocinétique , Humains , Préparations pharmaceutiques/métabolisme , Interactions médicamenteuses , Distribution tissulaire
11.
Chem Biol Drug Des ; 104(1): e14578, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39044291

RÉSUMÉ

The development of new radiopharmaceuticals for the detection of hidden infection foci has great relevance for early detection and the selection of the correct treatment, particularly in immunosuppressed patients. In that sense, the labelling of antimicrobial peptides (AMPs) that are capable of binding specifically to the pathogenic microorganism which causes the infection, should provide a sufficiently specific agent, able to distinguish an infection from a sterile inflammation. Defensins are particularly interesting molecules with antimicrobial activity, the EcgDf1 defensin was identified from the genome of a Uruguayan native plant, Erythrina crista-galli, the 'Ceibo' tree. Our group has previously reported a synthetic biologically active short analogue EcgDf21 (ERFTGGHCRGFRRRCFCTKHC) successfully labelled with 99mTc. Herein we present a shorter analogue which also preserves the γ-core domain, as a pharmacophore for a potential infection detection agent. This peptide was derivatized with the bifunctional chelating agent 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) through a lysine linker in the amino-terminal group (NOTA-KGHCRGFRRRC) and radiolabelled with 68Ga ([68Ga]Ga-NOTA-K-EcgDf1(10)). The [68Ga]Ga-NOTA-K-EcgDf1(10) labelling procedure rendered a product with high radiochemical purity and stability in the labelling milieu. The Log P value indicated that the complex has a hydrophilic behaviour, confirmed by the biodistribution profile. The [68Ga]Ga-NOTA-K-EcgDf1(10) complex demonstrated specific binding to cultures of Candida albicans and Aspergillus niger. Its biodistribution showed renal elimination and low accumulation in the rest of the body. It was possible to successfully differentiate sterile inflammation from infection by PET images in nude mice with a target/non-target ratio of 3.3 for C. albicans and 3.7 for A. niger, respectively.


Sujet(s)
Défensines , Radio-isotopes du gallium , Tomographie par émission de positons , Radiopharmaceutiques , Animaux , Humains , Souris , Séquence d'acides aminés , Défensines/composition chimique , Radio-isotopes du gallium/composition chimique , Peptides/composition chimique , Tomographie par émission de positons/méthodes , Radiopharmaceutiques/composition chimique , Distribution tissulaire , Composés organiques du technétium/composition chimique
12.
Recent Pat Anticancer Drug Discov ; 19(4): 503-515, 2024.
Article de Anglais | MEDLINE | ID: mdl-39044710

RÉSUMÉ

BACKGROUND: Both apurinic/apyrimidinic endodeoxyribonuclease 1 (APE1) inhibition and melatonin suppress prostate cancer (PCa) growth. OBJECTIVE: This study evaluated the therapeutic efficiency of self-assembled and prostate-specific membrane antigen (PSMA)-targeted nanocarrier loading 125I radioactive particles and encapsulating siRNA targeting APE1 (siAPE1) and melatonin for PCa. METHODS: The linear polyarginine R12 polypeptide was prepared using Fmoc-Arg-Pbf-OH. The PSMA-targeted polymer was synthesized by conjugating azide-modified R12 peptide to PSMA monoclonal antibody (mAb). Before experiments, the PSMA-R12 nanocarrier was installed with melatonin and siAPE1, which were subsequently labeled by 125I radioactive particles. In vitro biocompatibility and cytotoxicity of nanocomposites were examined in LNCaP cells and in vivo biodistribution and pharmacokinetics were determined using PCa tumor-bearing mice. RESULTS: PSMA-R12 nanocarrier was ~120 nm in size and was increased to ~150 nm by melatonin encapsulation. PSMA-R12 nanoparticles had efficient loading capacities of siAPE1, melatonin, and 125I particles. The co-delivery of melatonin and siAPE1 by PSMA-R12-125I showed synergistic effects on suppressing LNCaP cell proliferation and Bcl-2 expression and promoting cell apoptosis and caspase-3 expression. Pharmacokinetics analysis showed that Mel@PSMA-R12-125I particles had high uptake activity in the liver, spleen, kidney, intestine, and tumor, and were accumulated in the tumor sites within the first 8 h p.i., but was rapidly cleared from all the tested organs at 24 h p.i. Administration of nanoparticles to PCa tumors in vivo showed that Mel@PSMA-R12- 125I/siAPE1 had high efficiency in suppressing PCa tumor growth. CONCLUSION: The PSMA-targeted nanocarrier encapsulating siAPE1 and melatonin is a promising therapeutic strategy for PCa and can provide a theoretical basis for patent applications.


Sujet(s)
Antigènes de surface , Glutamate carboxypeptidase II , Radio-isotopes de l'iode , Mélatonine , Nanoparticules , Tumeurs de la prostate , Mâle , Animaux , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Humains , Radio-isotopes de l'iode/administration et posologie , Mélatonine/pharmacologie , Mélatonine/administration et posologie , Lignée cellulaire tumorale , Nanoparticules/composition chimique , Souris , Glutamate carboxypeptidase II/antagonistes et inhibiteurs , Glutamate carboxypeptidase II/métabolisme , Distribution tissulaire , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Apoptose/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Petit ARN interférent/administration et posologie , Petit ARN interférent/pharmacologie
13.
Drug Des Devel Ther ; 18: 2905-2917, 2024.
Article de Anglais | MEDLINE | ID: mdl-39011542

RÉSUMÉ

Background: Our previous studies in vitro and in vivo have shown anti-severe acute respiratory syndrome coronavirus 2 activity of fingerroot extract (Boesenbergia rotunda) and its phytochemical panduratin A. Aim of Study: Therefore, the objective of this study was to determine the pharmacokinetic profiles of panduratin A, as a pure compound and in fingerroot extract, in rats. Materials and Methods: Male rats were randomly divided into four groups. Rats underwent intravenous administration of 4.5 mg/kg panduratin A, a single oral administration of 45 mg/kg panduratin A, or a multiple oral administration of 45 mg/kg panduratin A-consisted fingerroot extract for 7 consecutive days. The concentrations of panduratin A in plasma, tissues, and excreta were measured by using LCMS with a validated method. Results: The rats showed no change in health status after receiving all test preparations. The absolute oral bioavailability of panduratin A administered as pure panduratin A and fingerroot extract were approximately 9% and 6%, respectively. The peak concentrations for the single oral doses of 45 mg/kg panduratin A and fingerroot extract, were 4833 ± 659 and 3269 ± 819 µg/L, respectively. Panduratin A was mostly distributed in gastrointestinal organs, with the highest tissue-to-plasma ratio in the stomach. Approximately 20-30% of unchanged panduratin A from the administered dose was detected in feces while a negligible amount was found in urine. The major metabolites of administered panduratin A were identified in feces as oxidation and dioxidation products. Conclusion: Panduratin A from fingerroot extract showed low oral bioavailability, good tissue distribution, and partially biotransformed before excretion via feces. These findings will assist in developing fingerroot extract as a phytopharmaceutical product for COVID-19 treatment.


Sujet(s)
Biodisponibilité , Extraits de plantes , Rat Sprague-Dawley , Zingiberaceae , Animaux , Mâle , Extraits de plantes/administration et posologie , Extraits de plantes/pharmacocinétique , Administration par voie orale , Rats , Zingiberaceae/composition chimique , Distribution tissulaire , Chalcones
15.
Sci Rep ; 14(1): 16141, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997436

RÉSUMÉ

Soluble biomarkers are paramount to personalized medicine. However, the in vivo turnover and biodistribution of soluble proteins is seldom characterized. The cleaved extracellular domain of the AXL receptor (sAXL) is a prognostic biomarker in several diseases and a predictive marker of AXL targeting agents. Plasma sAXL reflects a balance between production in tissues with lymphatic transport into the circulation and removal from blood by degradation or excretion. It is unclear how this transport cycle affects plasma sAXL levels that are the metric for biomarker development. Radiolabeled mouse sAxl was monitored after intravenous injection to measure degradation and urinary excretion of sAxl, and after intradermal injection to mimic tissue or tumor production. sAxl was rapidly taken-up and degraded by the liver and kidney cortex. Surprisingly, intact sAxl was detectable in urine, indicating passage through the glomerular filter and a unique sampling opportunity. The structure of sAxl showed an elongated, flexible molecule with a length of 18 nm and a thickness of only 3 nm, allowing passage through the glomerulus and excretion into the urine. Intradermally injected sAxl passed through local and distant lymph nodes, followed by uptake in liver and kidney cortex. Low levels of sAxl were seen in the plasma, consistent with an extended transit time from local tissue to circulation. The rapid plasma clearance of sAxl suggests that steady-state levels in blood will sensitively and dynamically reflect the rate of production of sAxl in the tissues but will be influenced by perturbations of liver and kidney function.


Sujet(s)
Axl Receptor Tyrosine Kinase , Marqueurs biologiques , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Animaux , Récepteurs à activité tyrosine kinase/métabolisme , Souris , Distribution tissulaire , Marqueurs biologiques/urine , Marqueurs biologiques/sang , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/urine , Protéines proto-oncogènes/sang , Foie/métabolisme , Humains , Femelle
16.
Molecules ; 29(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38999044

RÉSUMÉ

BACKGROUND: FAP radiopharmaceuticals show promise for cancer diagnosis; however, their limited tumor residency hinders treatment. This study compared two FAPi derivatives, DOTA.SA.FAPi and DOTAGA.(SA.FAPi)2, labeled with gallium-68 and lutetium-177, aiming to determine an optimum combination for creating theranostic pairs. METHODS: The radiotracers were studied for lipophilicity, binding to human serum proteins, and binding to human cancer-associated fibroblasts (CAFs) in vitro, including saturation and internalization/externalization studies. PET/SPECT/CT and biodistribution studies were conducted in PC3 and U87MG xenografts for [68Ga]Ga-DOTA.SA.FAPi and [68Ga]Ga-DOTAGA.(SA.FAPi)2. [177Lu]Lu-DOTA.SA.FAPi and [177Lu]Lu-DOTAGA.(SA.FAPi)2, were evaluated in PC3 xenografts. Biodistribution studies of [68Ga]Ga-DOTA.SA.FAPi were performed in healthy male and female mice. RESULTS: All radiotracers exhibited strong binding to FAP. Their internalization rate was fast while only [177Lu]Lu-DOTAGA.(SA.FAPi)2 was retained longer in CAFs. [68Ga]Ga-DOTAGA.(SA.FAPi)2 and [177Lu]Lu-DOTAGA.(SA.FAPi)2 displayed elevated lipophilicity and affinity for human serum proteins compared to [68Ga]Ga-DOTA.SA.FAPi and [177Lu]Lu-DOTA.SA.FAPi. In vivo studies revealed slower washout of [68Ga]Ga-DOTAGA.(SA.FAPi)2 within 3 h compared to [68Ga]Ga-DOTA.SA.FAPi. The tumor-to-tissue ratios of [68Ga]Ga-DOTAGA.(SA.FAPi)2 versus [68Ga]Ga-DOTA.SA.FAPi did not exhibit any significant differences. [177Lu]Lu-DOTAGA.(SA.FAPi)2 maintained a significant tumor uptake even after 96 h p.i. compared to [177Lu]Lu-DOTA.SA.FAPi. CONCLUSIONS: Dimeric compounds hold promise for therapy, while monomers are better suited for diagnostics. Finding the right combination is essential for effective disease management.


Sujet(s)
Endopeptidases , Radio-isotopes du gallium , Lutétium , Radio-isotopes , Radiopharmaceutiques , Lutétium/composition chimique , Humains , Animaux , Souris , Distribution tissulaire , Radio-isotopes/composition chimique , Radiopharmaceutiques/composition chimique , Radiopharmaceutiques/pharmacocinétique , Radiopharmaceutiques/pharmacologie , Radio-isotopes du gallium/composition chimique , Lignée cellulaire tumorale , Protéines membranaires/antagonistes et inhibiteurs , Protéines membranaires/métabolisme , Gelatinases/antagonistes et inhibiteurs , Gelatinases/métabolisme , Composés hétéromonocycliques/composition chimique , Femelle , Mâle , Nanomédecine théranostique
17.
Molecules ; 29(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38999054

RÉSUMÉ

Gastrin-releasing peptide receptor (GRPR), overexpressed in many solid tumors, is a promising imaging marker and therapeutic target. Most reported GRPR-targeted radioligands contain a C-terminal amide. Based on the reported potent antagonist D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHOH, we synthesized C-terminal hydroxamate-derived [68Ga]Ga-LW02075 ([68Ga]Ga-DOTA-pABzA-DIG-D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHOH) and [68Ga]Ga-LW02050 ([68Ga]Ga-DOTA-Pip-D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHOH), and compared them with the closely related and clinically validated [68Ga]Ga-SB3 ([68Ga]Ga-DOTA-pABzA-DIG-D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHEt). Binding affinities (Ki) of Ga-SB3, Ga-LW02075, and Ga-LW02050 were 1.20 ± 0.31, 1.39 ± 0.54, and 8.53 ± 1.52 nM, respectively. Both Ga-LW02075 and Ga-LW02050 were confirmed to be GRPR antagonists by calcium release assay. Imaging studies showed that PC-3 prostate cancer tumor xenografts were clearly visualized at 1 h post injection by [68Ga]Ga-SB3 and [68Ga]Ga-LW02050 in PET images, but not by [68Ga]Ga-LW02075. Ex vivo biodistribution studies conducted at 1 h post injection showed that the tumor uptake of [68Ga]Ga-LW02050 was comparable to that of [68Ga]Ga-SB3 (5.38 ± 1.00 vs. 6.98 ± 1.36 %ID/g), followed by [68Ga]Ga-LW02075 (3.97 ± 1.71 %ID/g). [68Ga]Ga-SB3 had the highest pancreas uptake (37.3 ± 6.90 %ID/g) followed by [68Ga]Ga-LW02075 (17.8 ± 5.24 %ID/g), while the pancreas uptake of [68Ga]Ga-LW02050 was only 0.53 ± 0.11 %ID/g. Our data suggest that [68Ga]Ga-LW02050 is a promising PET tracer for detecting GRPR-expressing cancer lesions.


Sujet(s)
Radio-isotopes du gallium , Acides hydroxamiques , Tomographie par émission de positons , Radiopharmaceutiques , Récepteur bombésine , Récepteur bombésine/métabolisme , Récepteur bombésine/antagonistes et inhibiteurs , Radio-isotopes du gallium/composition chimique , Animaux , Humains , Tomographie par émission de positons/méthodes , Souris , Acides hydroxamiques/composition chimique , Acides hydroxamiques/pharmacocinétique , Acides hydroxamiques/synthèse chimique , Radiopharmaceutiques/composition chimique , Radiopharmaceutiques/synthèse chimique , Radiopharmaceutiques/pharmacocinétique , Lignée cellulaire tumorale , Distribution tissulaire , Mâle , Tumeurs/imagerie diagnostique , Tumeurs/métabolisme , Tumeurs de la prostate/imagerie diagnostique , Tumeurs de la prostate/métabolisme
18.
Clin Pharmacokinet ; 63(7): 965-980, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38955946

RÉSUMÉ

BACKGROUND AND OBJECTIVE: The interstitial fluid of tissues is the effect site for antibiotics targeting extracellular pathogens. Microdialysis studies investigating these concentrations in muscle and subcutaneous tissue have reported notable variability in tissue penetration. This study aimed to comprehensively summarise the existing data on interstitial fluid penetration in these tissues and to identify potential factors influencing antibiotic distribution. METHODS: A literature review was conducted, focusing on subcutaneous and intramuscular microdialysis studies of antibiotics in both adult healthy volunteers and patients. Random-effect meta-analyses were used to aggregate effect size estimates of tissue penetration. The primary parameter of interest was the unbound penetration ratio, which represents the ratio of the area under the concentration-time curve in interstitial fluid relative to the area under the concentration-time curve in plasma, using unbound concentrations. RESULTS: In total, 52 reports were incorporated into this analysis. The unbound antibiotic exposure in the interstitial fluid of healthy volunteers was, on average, 22% lower than in plasma. The unbound penetration ratio values were higher after multiple dosing but did not significantly differ between muscle and subcutaneous tissue. Unbound penetration ratio values were lower for acids and bases compared with neutral antibiotics. Neither the molecular weight nor the logP of the antibiotics accounted for the variations in the unbound penetration ratio. Obesity was associated with lower interstitial fluid penetration. Conditions such as sepsis, tissue inflammation and tissue ischaemia were not significantly associated with altered interstitial fluid penetration. CONCLUSIONS: This study highlights the variability and generally lower exposure of unbound antibiotics in the subcutaneous and intramuscular interstitial fluid compared with exposure in plasma. Future research should focus on understanding the therapeutic relevance of these differences and identify key covariates that may influence them.


Sujet(s)
Antibactériens , Liquide extracellulaire , Microdialyse , Humains , Liquide extracellulaire/métabolisme , Liquide extracellulaire/composition chimique , Microdialyse/méthodes , Antibactériens/pharmacocinétique , Antibactériens/administration et posologie , Adulte , Tissu sous-cutané/métabolisme , Distribution tissulaire , Injections musculaires , Injections sous-cutanées
19.
Anal Chem ; 96(29): 11725-11733, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38975941

RÉSUMÉ

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.


Sujet(s)
Radio-isotopes du fluor , Imagerie de perfusion myocardique , Tomographie par émission de positons , Radiopharmaceutiques , Animaux , Radio-isotopes du fluor/composition chimique , Imagerie de perfusion myocardique/méthodes , Souris , Radiopharmaceutiques/composition chimique , Distribution tissulaire , Humains , Mâle , Ischémie myocardique/imagerie diagnostique
20.
Int J Nanomedicine ; 19: 6717-6730, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979530

RÉSUMÉ

Introduction: Immune regulatory small molecule JQ1 can block its downstream effector PD-L1 pathway and effectively reverse the PD-L1 upregulation induced by doxorubicin (DOX). So the synergistic administration of chemotherapeutic drug DOX and JQ1 is expected to increase the sensitivity of tumors to immune checkpoint therapy and jointly enhance the body's own immunity, thus effectively killing tumor cells. Therefore, a drug delivery system loaded with DOX and JQ1 was devised in this study. Methods: Polydopamine nanoparticles (PDA NPs) were synthesized through spontaneous polymerization. Under appropriate pH conditions, DOX and JQ1 were loaded onto the surface of PDA NPs, and the release of DOX and JQ1 were measured using UV-Vis or high performance liquid chromatography (HPLC). The mechanism of fabricated nanocomplex in vitro was investigated by cell uptake experiment, cell viability assays, apoptosis assays, and Western blot analysis. Finally, the tumor-bearing mouse model was used to evaluate the tumor-inhibiting efficacy and the biosafety in vivo. Results: JQ1 and DOX were successfully loaded onto PDA NPs. PDA-DOX/JQ1 NPs inhibited the growth of prostate cancer cells, reduced the expression of apoptosis related proteins and induced apoptosis in vitro. The in vivo biodistribution indicated that PDA-DOX/JQ1 NPs could accumulated at the tumor sites through the EPR effect. In tumor-bearing mice, JQ1 delivered with PDA-DOX/JQ1 NPs reduced PD-L1 expression at tumor sites, generating significant tumor suppression. Furthermore, PDA-DOX/JQ1 NPs could reduce the side effects, and produce good synergistic treatment effect in vivo. Conclusion: We have successfully prepared a multifunctional platform for synergistic prostate cancer therapy.


Sujet(s)
Apoptose , Azépines , Doxorubicine , Indoles , Nanoparticules , Polymères , Tumeurs de la prostate , Mâle , Animaux , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Doxorubicine/pharmacocinétique , Doxorubicine/administration et posologie , Indoles/composition chimique , Indoles/pharmacologie , Indoles/pharmacocinétique , Polymères/composition chimique , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Nanoparticules/composition chimique , Humains , Souris , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Azépines/composition chimique , Azépines/pharmacologie , Azépines/pharmacocinétique , Synergie des médicaments , Survie cellulaire/effets des médicaments et des substances chimiques , Distribution tissulaire , Tests d'activité antitumorale sur modèle de xénogreffe , Libération de médicament , Vecteurs de médicaments/composition chimique , Vecteurs de médicaments/pharmacocinétique , Antigène CD274/métabolisme , Triazoles
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE