Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.056
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 183(3): 786-801.e19, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33125893

RESUMEN

Trained immunity, a functional state of myeloid cells, has been proposed as a compelling immune-oncological target. Its efficient induction requires direct engagement of myeloid progenitors in the bone marrow. For this purpose, we developed a bone marrow-avid nanobiologic platform designed specifically to induce trained immunity. We established the potent anti-tumor capabilities of our lead candidate MTP10-HDL in a B16F10 mouse melanoma model. These anti-tumor effects result from trained immunity-induced myelopoiesis caused by epigenetic rewiring of multipotent progenitors in the bone marrow, which overcomes the immunosuppressive tumor microenvironment. Furthermore, MTP10-HDL nanotherapy potentiates checkpoint inhibition in this melanoma model refractory to anti-PD-1 and anti-CTLA-4 therapy. Finally, we determined MTP10-HDL's favorable biodistribution and safety profile in non-human primates. In conclusion, we show that rationally designed nanobiologics can promote trained immunity and elicit a durable anti-tumor response either as a monotherapy or in combination with checkpoint inhibitor drugs.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunidad , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Nanotecnología , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Animales , Conducta Animal , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Proliferación Celular/efectos de los fármacos , Colesterol/metabolismo , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunidad/efectos de los fármacos , Inmunoterapia , Lipoproteínas HDL/metabolismo , Ratones Endogámicos C57BL , Primates , Distribución Tisular/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
2.
Toxicol Appl Pharmacol ; 434: 115796, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34785274

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has required the urgent development of new therapies, among which passive immunotherapy is contemplated. CoviFab (INM005) is a RBD-specific F(ab')2 fragment derived from equine polyclonal antibodies. We investigate their preclinical security and biodistribution by in vivo and ex vivo NIR imaging after intravenous administration of a dose of 4 mg/kg at time 0 and 48 h. Images were taken at 1, 12, 24, 36, 48, 49, 60, 72, 84, 96, 108, 120, 132 and 144 h after the first intravenous injection. At 96 and 144 h, mice were sacrificed for haematology, serum chemistry, clinical pathology, histopathology and ex vivo imaging. The biodistribution profile was similar in all organs studied, with the highest fluorescence at 1 h after each injection, gradually decreasing after that each one and until the end of the study (144 h). The toxicology study revealed no significant changes in the haematology and serum chemistry parameters. Further, there were no changes in the gross and histological examination of organs. Nonclinical data of the current study confirm that CoviFab is safe, without observable adverse effects in mice. Furthermore, we confirm that bioimaging studies are a useful approach in preclinical trials to determine biodistribution.


Asunto(s)
Anticuerpos Antivirales/metabolismo , Tratamiento Farmacológico de COVID-19 , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes/metabolismo , SARS-CoV-2/metabolismo , Administración Intravenosa , Animales , Anticuerpos Antivirales/administración & dosificación , Anticuerpos Antivirales/efectos adversos , COVID-19/metabolismo , COVID-19/prevención & control , Células HEK293 , Caballos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Receptores Inmunológicos/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , SARS-CoV-2/efectos de los fármacos , Espectroscopía Infrarroja Corta/métodos , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
3.
Am J Physiol Endocrinol Metab ; 320(3): E641-E652, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33427052

RESUMEN

Pegylated arginine deiminase (ADI-PEG20) results in the depletion of arginine with the production of isomolar amounts of citrulline. This citrulline has the potential to be utilized by the citrulline recycling pathway regenerating arginine and sustaining tissue arginine availability. The goal of this research was to test the hypothesis that ADI-PEG20 depletes circulating arginine in pigs but maintains tissue arginine concentration and function, and to characterize the kinetics of citrulline and arginine. Two multitracer approaches (bolus dose and primed-continuous infusion) were used to investigate the metabolism of arginine and citrulline in Control (n = 7) and ADI-PEG20 treated (n = 8) pigs during the postprandial period. In addition, blood pressure was monitored by telemetry, and multiple tissues were collected to determine arginine concentration. Plasma arginine was depleted immediately after ADI-PEG20 administration, with an increase in plasma citrulline concentration (P < 0.01). The depletion of arginine did not affect (P > 0.10) blood pressure, whole body protein synthesis, or urea production. Despite the lack of circulating arginine in ADI-PEG20-treated pigs, most tissues were able to maintain concentrations similar (P > 0.10) to those in Control animals. The kinetics of citrulline and arginine indicated the high citrulline turnover and regeneration of arginine through the citrulline recycling pathway. ADI-PEG20 administration resulted in an absolute and almost instantaneous depletion of circulating arginine, thus reducing global availability without affecting cardiovascular parameters and protein metabolism. The citrulline produced from the deimination of arginine was in turn utilized by the citrulline recycling pathway restoring local tissue arginine availability.NEW & NOTEWORTHY Pegylated arginine deiminase depletes circulating arginine, but the citrulline generated is utilized by multiple tissues to regenerate arginine and sustain local arginine availability. Preempting the arginine depletion that occurs as result of sepsis and trauma with arginine deiminase offers the possibility of maintaining tissue arginine availability despite negligible plasma arginine concentrations.


Asunto(s)
Arginina/sangre , Arginina/farmacocinética , Hidrolasas/farmacología , Polietilenglicoles/farmacología , Animales , Disponibilidad Biológica , Citrulina/sangre , Femenino , Cinética , Masculino , Porcinos , Distribución Tisular/efectos de los fármacos
4.
J Pharmacol Exp Ther ; 379(2): 134-146, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34413198

RESUMEN

Conjugation of small interfering RNA (siRNA) to tris N-acetylgalactosamine [(GalNAc)3] can enable highly selective, potent, and durable knockdown of targeted proteins in the liver. However, potential knowledge gaps between in vitro experiments, preclinical species, and clinical scenarios remain. A minimal physiologically based pharmacokinetic-pharmacodynamic model for GalNAc-conjugated siRNA (GalNAc-siRNA) was developed using published data for fitusiran (ALN-AT3), an investigational compound targeting liver antithrombin (AT), to delineate putative determinants governing the whole-body-to-cellular pharmacokinetic (PK) and pharmacodynamic (PD) properties of GalNAc-siRNA and facilitate preclinical-to-clinical translation. The model mathematically linked relevant mechanisms: 1) hepatic biodistribution, 2) tris-GalNAc binding to asialoglycoprotein receptors (ASGPRs) on hepatocytes, 3) ASGPR endocytosis and recycling, 4) endosomal transport and escape of siRNA, 5) cytoplasmic RNA-induced silencing complex (RISC) loading, 6) degradation of target mRNA by bound RISC, and 7) knockdown of protein. Physiologic values for 36 out of 48 model parameters were obtained from the literature. Kinetic parameters governing (GalNAc)3-ASGPR binding and internalization were derived from published studies of uptake in hepatocytes. The proposed model well characterized reported pharmacokinetics, RISC dynamics, and knockdown of AT mRNA and protein by ALN-AT3 in mice. The model bridged multiple PK-PD data sets in preclinical species (mice, rat, monkey) and successfully captured reported plasma pharmacokinetics and AT knockdown in a phase I ascending-dose study. Estimates of in vivo potency were similar (∼2-fold) across species. Subcutaneous absorption and serum AT degradation rate constants scaled across species by body weight with allometric exponents of -0.29 and -0.22. The proposed mechanistic modeling framework characterizes the unique PK-PD properties of GalNAc-siRNA. SIGNIFICANCE STATEMENT: Tris N-acetylgalactosamine (GalNAc)3-conjugated small interfering RNA (siRNA) therapeutics enable liver-targeted gene therapy and precision medicine. Using a translational and systems-based minimal physiologically based pharmacokinetic-pharmacodynamic (mPBPK-PD) modeling approach, putative determinants influencing GalNAc-conjugated siRNA (GalNAc-siRNA) functionality in three preclinical species and humans were investigated. The developed model successfully integrated and characterized relevant published in vitro-derived biomeasures, mechanistic PK-PD profiles in animals, and observed clinical PK-PD responses for an investigational GalNAc-siRNA (fitusiran). This modeling effort delineates the disposition and liver-targeted pharmacodynamics of GalNAc-siRNA.


Asunto(s)
Acetilgalactosamina/farmacocinética , Silenciador del Gen/fisiología , Modelos Biológicos , ARN Interferente Pequeño/farmacocinética , Acetilgalactosamina/genética , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Haplorrinos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ratones , ARN Interferente Pequeño/genética , Ratas , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
5.
Drug Metab Dispos ; 49(6): 434-441, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33762296

RESUMEN

Disulfiram, an antialcoholism drug, could potentially be repurposed as an anticancer drug because of the formation of copper(II) diethyldithiocarbamate (CuET) from dithiocarb (DTC, a reduced metabolite of disulfiram) and Cu2+ CuET exhibited preferential distribution to tumor tissues. This study investigated the mechanism of CuET accumulation in tumor tissues by employing MDA-MB-231 human breast cancer cells. The concentration of CuET in cells treated with DTC and Cu2+ in acidic culture medium (pH 6.8) was significantly higher than that of the control group (pH 7.4). Subsequently, the effects of pH on the uptake of DTC, Cu2+, and CuET were investigated separately. The acidic environment significantly increased the uptake rate of DTC and Cu2+ but had no effect on CuET. MDA-MB-231 cells overexpressing copper transporter hCTR1 were constructed to evaluate its intermediate role in CuET accumulation. After treatment with CuCl2 followed by DTC for 15 minutes, the levels of CuET and Cu2+ in hCTR1-overexpressed cells were 2.5 times as much as those of vector group. In the tumors of cancer xenograft models constructed by hCTR1-MDA-MB-231 cells, the concentrations of CuET and Cu were also significantly higher than those of control group. In conclusion, the acidic microenvironment of tumors can promote the enrichment of CuET in tumors through dual action. On the one hand, it can promote transmembrane transport of DTC by converting ionic DTC into molecular state. On the other hand, it enhances Cu2+ uptake by activating hCTR1, which ultimately leads to the enrichment of CuET. SIGNIFICANCE STATEMENT: Increasing evidence suggests that the antitumor activity of disulfiram is related to the formation of a copper(II) diethyldithiocarbamate (CuET) of its reducing metabolite dithiocarb with copper(II) ion, which is preferentially distributed in tumor tissues. We showed that the acidic microenvironment, a common feature of many solid tumor tissues, could promote intracellular CuET accumulation through dual action without changing CuET uptake. This result is helpful for the formulation of clinical dosage regimens of disulfiram in cancer treatment.


Asunto(s)
Disulfiram/farmacología , Neoplasias , Distribución Tisular , Microambiente Tumoral , Inhibidores del Acetaldehído Deshidrogenasa/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Cobre/metabolismo , Transportador de Cobre 1/metabolismo , Reposicionamiento de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología , Oligoelementos/farmacología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología
6.
Drug Metab Dispos ; 49(4): 330-336, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33531412

RESUMEN

Volume of distribution (Vd) is a primary pharmacokinetic parameter used to calculate the half-life and plasma concentration-time profile of drugs. Numerous models have been relatively successful in predicting Vd, but the model developed by Korzekwa and Nagar is of particular interest because it utilizes plasma protein binding and microsomal binding data, both of which are readily available in vitro parameters. Here, Korzekwa and Nagar's model was validated and expanded upon using external and internal data sets. Tissue binding, plasma protein binding, Vd, physiochemical, and physiologic data sets were procured from literature and Genentech's internal data base. First, we investigated the hypothesis that tissue binding is primarily governed by passive processes that depend on the lipid composition of the tissue type. The fraction unbound in tissues (futissue) was very similar across human, rat, and mouse. In addition, we showed that dilution factors could be generated from nonlinear regression so that one futissue value could be used to estimate another one regardless of species. More importantly, results suggested that microsomes could serve as a surrogate for tissue binding. We applied the parameters from Korzekwa and Nagar's Vd model to two distinct liver microsomal data sets and found remarkably close statistical results. Brain and lung data sets also accurately predicted Vd, further validating the model. Vd prediction accuracy for compounds with log D7.4 > 1 significantly outperformed that of more hydrophilic compounds. Finally, human Vd predictions from Korzekwa and Nagar's model appear to be as accurate as rat allometry and slightly less accurate than dog and cynomolgus allometry. SIGNIFICANCE STATEMENT: This study shows that tissue binding is comparable across five species and can be interconverted with a dilution factor. In addition, we applied internal and external data sets to the volume of distribution model developed by Korzekwa and Nagar and found comparable Vd prediction accuracy between the Vd model and single-species allometry. These findings could potentially accelerate the drug research and development process by reducing the amount of resources associated with in vitro binding and animal experiments.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Encéfalo/metabolismo , Pulmón/metabolismo , Microsomas Hepáticos/metabolismo , Preparaciones Farmacéuticas/metabolismo , Distribución Tisular/fisiología , Animales , Encéfalo/efectos de los fármacos , Bases de Datos Factuales , Perros , Predicción , Humanos , Pulmón/efectos de los fármacos , Macaca fascicularis , Ratones , Microsomas Hepáticos/efectos de los fármacos , Preparaciones Farmacéuticas/administración & dosificación , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Ratas , Especificidad de la Especie , Distribución Tisular/efectos de los fármacos
7.
Toxicol Appl Pharmacol ; 428: 115679, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34400196

RESUMEN

It is known that, as the vast majority of the anthropogenically emitted mercury can be found in aquatic ecosystems, where several methylating bacteria are present, fish consumption represents the most critical intake source of the most toxic form of mercury, the methylmercury (MeHg). The aim of this work is to predict MeHg levels in the fish muscles which, being the edible portion, are part of the human diet. A physiologically based toxicokinetics model was developed to evaluate the kinetics of MeHg in red mullets. Fishes were described by means of a multi-compartment model including stomach, gut, blood, muscles and an additional compartment virtually encompassing all the remaining organs. Absorption, distribution and excretion were modelled considering different MeHg routes of administration and excretion: intake by ingestion of contaminated food, intake and elimination through inhalation-exhalation and excretion through feces. The model has been firstly validated on Terapon jarbua fish (using the weighted least squares method for parameter estimation) to be subsequently readapted to predict methylmercury concentrations in the muscle of red mullets (using an approximate Bayesian computation approach). This simple multicompartmental model could be considered part, a link in the chain, of a wider more complex project aiming at tracking the fate of MeHg from polluted seawater to the human end consumer. The present study could be useful to surveillance organizations in order to carry out a more comprehensive and informed risk assessment analysis and to take appropriate preventive measures by evaluating possible new MeHg concentration thresholds to minimize public health hazards.


Asunto(s)
Compuestos de Metilmercurio/farmacocinética , Compuestos de Metilmercurio/toxicidad , Smegmamorpha/metabolismo , Contaminantes Químicos del Agua/farmacocinética , Contaminantes Químicos del Agua/toxicidad , Animales , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología , Toxicocinética
8.
Mol Pharm ; 18(3): 1102-1120, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33356314

RESUMEN

Hepatocellular carcinoma (HCC) is a major cause of concern as it has substantial morbidity associated with it. Previous reports have ascertained the antiproliferative activity of imatinib mesylate (IMS) against diverse types of carcinomas, but limited bioavailability has also been reported. The present study envisaged optimized IMS-loaded lactoferrin (LF)-modified PEGylated liquid crystalline nanoparticles (IMS-LF-LCNPs) for effective therapy of IMS to HCC via asialoglycoprotein receptor (ASGPR) targeting. Results displayed that IMS-LF-LCNPs presented an optimum particle size of 120.40 ± 2.75 nm, a zeta potential of +12.5 ± 0.23 mV, and 73.94 ± 2.69% release. High-resolution transmission electron microscopy and atomic force microscopy were used to confirm the surface architecture of IMS-LF-LCNPs. The results of cytotoxicity and 4,6-diamidino-2-phenylindole revealed that IMS-LF-LCNPs had the highest growth inhibition and significant apoptotic effects. Pharmacokinetics and biodistribution studies showed that IMS-LF-LCNPs have superior pharmacokinetic performance and targeted delivery compared to IMS-LCNPs and plain IMS, which was attributed to the targeting action of LF that targets the ASGPR in hepatic cells. Next, our in vivo experiment established that the HCC environment existed due to suppression of BAX, cyt c, BAD, e-NOS, and caspase (3 and 9) genes, which thus owed upstream expression of Bcl-xl, iNOS, and Bcl-2 genes. The excellent therapeutic potential of IMS-LF-LCNPs began the significant stimulation of caspase-mediated apoptotic signals accountable for its anti-HCC prospect. 1H nuclear magnetic resonance (serum) metabolomics revealed that IMS-LF-LCNPs are capable of regulating the disturbed levels of metabolites linked to HCC triggered through N-nitrosodiethylamine. Therefore, IMS-LF-LCNPs are a potentially effective formulation against HCC.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/dietoterapia , Mesilato de Imatinib/farmacología , Lactoferrina/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Nanopartículas/química , Animales , Disponibilidad Biológica , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Células Hep G2 , Humanos , Cristales Líquidos/química , Neoplasias Hepáticas/genética , Masculino , Mitocondrias/genética , Tamaño de la Partícula , Polietilenglicoles/química , Ratas , Ratas Wistar , Distribución Tisular/efectos de los fármacos
9.
Mol Pharm ; 18(3): 1419-1430, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33522827

RESUMEN

The incidence and progression of inflammatory bowel disease are closely related to oxidative stress caused by excessive production of reactive oxygen species (ROS). To develop an efficacious and safe nanotherapy against inflammatory bowel diseases (IBD), we designed a novel pH/ROS dual-responsive prodrug micelle GC-B-Que as an inflammatory-targeted drug, which was comprised by active quercetin (Que) covalently linked to biocompatible glycol chitosan (GC) by aryl boronic ester as a responsive linker. The optimized micelles exhibited well-controlled physiochemical properties and stability in a physiological environment. Time-dependent NMR spectra traced the changes in the polymer structure in the presence of H2O2, confirming the release of the drug. The in vitro drug release studies indicated a low release rate (<20 wt %) in physiological conditions, but nearly complete release (>95 wt % after 72 h incubation) in a pH 5.8 medium containing 10 µM H2O2, exhibiting a pH/ROS dual-responsive property and sustained release behavior. Importantly, the negligible drug release in a simulated gastric environment in 1 h allowed us to perform intragastric administration, which has potential to achieve the oral delivery by mature enteric-coating modification in future. Further in vivo activities and biodistribution experiments found that the GC-B-Que micelles tended to accumulate in intestinal inflammation sites and showed better therapeutic efficacy than the free drugs (quercetin and mesalazine) in a colitis mice model. Typical inflammatory cytokines including TNF-α, IL-6, and iNOS were significantly suppressed by GC-B-Que micelle treatment. Our work promoted inflammatory-targeted delivery and intestinal drug accumulation for active single drug quercetin and improved the therapeutic effect of IBD. The current study also provided an alternative strategy for designing a smart responsive nanocarrier for a catechol-based drug to better achieve the target drug delivery.


Asunto(s)
Quitosano/química , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Profármacos/química , Profármacos/farmacología , Quercetina/química , Animales , Células CACO-2 , Línea Celular Tumoral , Citocinas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos/efectos de los fármacos , Células HT29 , Humanos , Peróxido de Hidrógeno/farmacología , Concentración de Iones de Hidrógeno , Enfermedades Inflamatorias del Intestino/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Micelas , Nanopartículas/química , Polímeros/química , Especies Reactivas de Oxígeno/metabolismo , Distribución Tisular/efectos de los fármacos
10.
Mol Pharm ; 18(1): 187-197, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33253591

RESUMEN

C-X-C chemokine receptor 4 (CXCR4) is highly expressed in cancers, contributing to proliferation, metastasis, and a poor prognosis. The noninvasive imaging of CXCR4 can enable the detection and characterization of aggressive cancers with poor outcomes. Currently, no 18F-labeled CXCR4 positron emission tomography (PET) radiotracer has demonstrated imaging contrast comparable to [68Ga]Ga-Pentixafor, a CXCR4-targeting radioligand. We, therefore, aimed to develop a high-contrast CXCR4-targeting radiotracer by incorporating a hydrophilic linker and trifluoroborate radioprosthesis to LY2510924, a known CXCR4 antagonist. A carboxy-ammoniomethyl-trifluoroborate (PepBF3) moiety was conjugated to the LY2510924-derived peptide possessing a triglutamate linker via amide bond formation to obtain BL08, whereas an alkyne ammoniomethyl-trifluoroborate (AMBF3) moiety was conjugated using the copper-catalyzed [3+2] cycloaddition click reaction to obtain BL09. BL08 and BL09 were radiolabeled with [18F]fluoride ion using 18F-19F isotope exchange. Pentixafor was radiolabeled with [68Ga]GaCl3. Side-by-side PET imaging and biodistribution studies were performed on immunocompromised mice bearing Daudi Burkitt lymphoma xenografts. The biodistribution of [18F]BL08 and [18F]BL09 showed tumor uptake at 2 h postinjection (p.i.) (5.67 ± 1.25%ID/g and 5.83 ± 0.92%ID/g, respectively), which were concordant with the results of PET imaging. [18F]BL08 had low background activity, providing tumor-to-blood, -muscle, and -liver ratios of 72 ± 20, 339 ± 81, and 14 ± 3 (2 h p.i.), respectively. [18F]BL09 behaved similarly, with ratios of 64 ± 20, 239 ± 72, and 17 ± 3 (2 h p.i.), respectively. This resulted in high-contrast visualization of tumors on PET imaging for both radiotracers. [18F]BL08 exhibited lower kidney uptake (2.2 ± 0.5%ID/g) compared to [18F]BL09 (7.6 ± 1.0%ID/g) at 2 h p.i. [18F]BL08 and [18F]BL09 demonstrated higher tumor-to-blood, -muscle, and -liver ratios compared to [68Ga]Ga-Pentixafor (18.9 ± 2.7, 95.4 ± 36.7, and 5.9 ± 0.7 at 2 h p.i., respectively). In conclusion, [18F]BL08 and [18F]BL09 enable high-contrast visualization of CXCR4 expression in Daudi xenografts. Based on high tumor-to-organ ratios, [18F]BL08 may prove a valuable new tool for CXCR4-targeted PET imaging with potential for translation. The use of a PepBF3 moiety is a new approach for the orthogonal conjugation of organotrifluoroborates for 18F-labeling of peptides.


Asunto(s)
Fluoruros/metabolismo , Radioisótopos de Flúor/metabolismo , Tomografía de Emisión de Positrones/métodos , Radiofármacos/metabolismo , Receptores CXCR4/metabolismo , Animales , Células CHO , Línea Celular , Complejos de Coordinación/metabolismo , Cricetulus , Masculino , Ratones , Ratones Endogámicos NOD , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Péptidos/metabolismo , Péptidos Cíclicos/metabolismo , Distribución Tisular/efectos de los fármacos
11.
Mol Pharm ; 18(6): 2360-2374, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34027660

RESUMEN

With a poor prognosis, glioblastoma multiforme is the most aggressive tumor of the central nervous system in humans. The aim of this study was to develop novel tracers for the tumor targeting and imaging of overexpressed serotonin-7 receptors (5-HT7Rs) in U-87 MG glioma xenografted nude mice. Two phenylpiperazine derivatives named as PHH and MPHH were designed, and the corresponding radiotracers 99mTc-PHH and 99mTc-MPHH were synthesized in high radiochemical purity (>95%). 99mTc-MPHH showed a higher affinity to 5-HT7Rs on U-87 MG cells compared to 99mTc-PHH. In biodistribution studies, the radiocomplexes showed good brain uptake at 15 min combined with good radioactivity retention in the brain for 240 min. Regional rabbit brain studies indicated a higher radioactivity concentration in the hippocampus and diencephalon than in the cerebellum. Compared to 99mTc-MPHH, the 99mTc-PHH exhibited a significantly increased tumor uptake at 15 and 60 min, but the rapid blood clearance of 99mTc-MPHH led to enhanced tumor-to-muscle ratios at 240 min. A significant reduction in tumor uptake 60 min after an injection of pimozide (5-HT7 receptor antagonist) confirms the tumor uptake was receptor-mediated specifically. The tumor-to-contralateral muscle tissue ratio of 99mTc-PHH and 99mTc-MPHH in nude mice with U-87 MG xenograft was measured (5.25 and 4.65) at 60 min as well as (6.25 and 6.76) at 240 min, respectively.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Glioblastoma/diagnóstico por imagen , Piperazinas/administración & dosificación , Radiofármacos/administración & dosificación , Receptores de Serotonina/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/patología , Humanos , Ligandos , Masculino , Ratones , Pimozida/administración & dosificación , Piperazinas/síntesis química , Piperazinas/farmacocinética , Tomografía de Emisión de Positrones/métodos , Conejos , Radiofármacos/síntesis química , Radiofármacos/farmacocinética , Antagonistas de la Serotonina/administración & dosificación , Tecnecio , Distribución Tisular/efectos de los fármacos , Tomografía Computarizada de Emisión de Fotón Único/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Mol Pharm ; 18(8): 3026-3036, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34213912

RESUMEN

Breast cancer is one of the most common malignant tumors in women. The existence of multiple breast cancer subtypes often leads to chemotherapy failure or the development of drug resistance. In recent years, photodynamic therapy has been proven to enhance the sensitivity of tumors to chemotherapeutic drugs. Porphyrin-based metal-organic framework (MOF) materials could simultaneously be used as carriers for chemotherapy and photosensitizers in photodynamic therapy. In this paper, doxorubicin hydrochloride (DOX) was loaded in porphyrin MOFs, and the mechanism of the synergistic effect of the DOX carriers and photodynamic therapy on breast cancer was investigated. In vitro and in vivo experiments have shown that MOFs could prolong the residence time of DOX in tumor tissues and promote the endocytosis of DOX by tumor cells. In addition, adjuvant treatment with photodynamic therapy can promote breast cancer tumors to resensitize to DOX and synergistically enhance the chemotherapy effect of DOX. Therefore, this study can provide effective development ideas for reversing drug resistance during breast cancer chemotherapy and improving the therapeutic effect of chemotherapy on breast cancer.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Doxorrubicina/administración & dosificación , Nanopartículas del Metal/química , Estructuras Metalorgánicas/química , Sistema de Administración de Fármacos con Nanopartículas/química , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/química , Animales , Antibióticos Antineoplásicos/farmacocinética , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Doxorrubicina/farmacocinética , Liberación de Fármacos , Endocitosis/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Porfirinas/química , Distribución Tisular/efectos de los fármacos , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos
13.
Anesthesiology ; 135(4): 724-727, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34499097

RESUMEN

The Solubility of Halothane in Blood and Tissue Homogenates. By Larson CP, Eger EI, Severinghaus JW. Anesthesiology 1962; 23:349-55. Measured samples of human and bovine blood, human hemoglobin, and tissue homogenates from human fat and both human and bovine liver, kidney, muscle, whole brain, and separated gray and white cortex were added to stoppered 2,000-ml Erlenmeyer flasks. To each flask, 0.1 ml of liquid halothane was added under negative pressure using a calibrated micropipette. After the flask was agitated for 2 to 4 h to achieve equilibrium between the gas and blood or tissue contents, a calibrated infrared halothane analyzer was used to measure the concentration of halothane vapor. Calculated partition coefficients ranged from 0.7 for water to 2.3 for blood and from 3.5 for human or bovine kidney to 6 for human whole brain or liver and 8 for human muscle. Human peritoneal fat had a value of 138. The human blood-gas partition coefficient of 2.3 as determined by this equilibration method was well below the previously published value of 3.6.


Asunto(s)
Anestésicos por Inhalación/metabolismo , Investigación Biomédica/normas , Halotano/metabolismo , Anestésicos por Inhalación/química , Animales , Bovinos , Halotano/química , Humanos , Solubilidad/efectos de los fármacos , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
14.
Mol Biol Rep ; 48(5): 4591-4600, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34125331

RESUMEN

Our previous study showed that soy milks could contain high levels of active soybean trypsin inhibitors (SBTI) if they were not properly processed. This study investigated the effects of consuming active SBTI on pancreatic weights, histology, trypsinogen production and expression of STAT3, receptors for androgen (AR) and estrogen (ER) in pancreas, liver and uterus of rats. Weanling Sprague-Dawley rats were randomly divided into 3 groups (8 females and 8 males/group) and fed diets containing either 20% casein protein (Casein) or 20% soy protein (SP) in the presence of high (1.42 BAEE unit/µg, SP + SBTI) or low (0.2 BAEE unit/µg, SP-SBTI) levels of active SBTI for 8 weeks. Ingestion of SP + SBTI diet markedly increased pancreatic weights and trypsinogen content (p < 0.01), and caused acinar cell hypertrophy, and reduced pancreatic STAT3, p-STAT3, AR and ERß content, and increased uterine ERα and ERß compared to the Casein or SP-SBTI diets (p < 0.05). The two SP-containing diets lowered hepatic STAT3, p-STAT3, and pancreatic ERα, and increased hepatic ERα and ERß content in the female rats compared to the Casein diet (p < 0.05). This study demonstrated for the first time that consumption of high level of active SBTI not only increased pancreatic weights and acinar cell secretions, but also attenuated the expression of pancreatic STAT3, p-STAT3, AR, and ERß proteins in both sexes and increased uterine ERα and ERß content, and that dietary soy protein affected hepatic STAT3, p-STAT3, ERα and ERß in a gender-dependent manner.


Asunto(s)
Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Páncreas/metabolismo , Factor de Transcripción STAT3/genética , Inhibidores de Tripsina/farmacología , Animales , Estrógenos/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hígado , Masculino , Páncreas/efectos de los fármacos , Ratas , Receptores Androgénicos/genética , Proteínas de Soja/genética , Proteínas de Soja/farmacología , Glycine max/química , Distribución Tisular/efectos de los fármacos , Distribución Tisular/genética , Pesos y Medidas/normas
15.
Anesth Analg ; 132(1): 110-118, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32118620

RESUMEN

BACKGROUND: Propofol can be measured in exhaled gas. Exhaled and plasma propofol concentrations correlate well, but the relationship with tissue concentrations remains unknown. We thus evaluated the relationship between exhaled, plasma, and various tissue propofol concentrations. Because the drug acts in the brain, we focused on the relationship between exhaled and brain tissue propofol concentrations. METHODS: Thirty-six male Sprague-Dawley rats were anesthetized with propofol, ketamine, and rocuronium for 6 hours. Animals were randomly assigned to propofol infusions at 20, 40, or 60 mg·kg·h (n = 12 per group). Exhaled propofol concentrations were measured at 15-minute intervals by multicapillary column-ion mobility spectrometry. Arterial blood samples, 110 µL each, were collected 15, 30, and 45 minutes, and 1, 2, 4, and 6 hours after the propofol infusion started. Propofol concentrations were measured in brain, lung, liver, kidney, muscle, and fat tissue after 6 hours. The last exhaled and plasma concentrations were used for linear regression analyses with tissue concentrations. RESULTS: The correlation of exhaled versus plasma concentrations (R = 0.71) was comparable to the correlation of exhaled versus brain tissue concentrations (R = 0.75) at the end of the study. In contrast, correlations between plasma and lung and between lung and exhaled propofol concentrations were poor. Less than a part-per-thousand of propofol was exhaled over 6 hours. CONCLUSIONS: Exhaled propofol concentrations correlate reasonably well with brain tissue and plasma concentrations in rats, and may thus be useful to estimate anesthetic drug effect. The equilibration between plasma propofol and exhaled gas is apparently independent of lung tissue concentration. Only a tiny fraction of administered propofol is eliminated via the lungs, and exhaled quantities thus have negligible influence on plasma concentrations.


Asunto(s)
Anestésicos Intravenosos/metabolismo , Encéfalo/metabolismo , Plasma/metabolismo , Propofol/metabolismo , Anestésicos Intravenosos/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Pruebas Respiratorias/métodos , Espiración/efectos de los fármacos , Masculino , Plasma/efectos de los fármacos , Propofol/administración & dosificación , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
16.
Biol Pharm Bull ; 44(1): 1-6, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390535

RESUMEN

Protein affinity reagents are widely used for basic research, diagnostics, and disease therapy. Antibodies and their fragments are known as the most common protein affinity reagents. They specifically and strongly bind to target molecules and inhibit their functions. Thus, antibody drugs have increased in the recent two decades for disease therapy, such as cancer. These strong protein-protein interactions are composed of a nexus of multiple weak interactions. Synthetic polymers that bind to target molecules have been developed by the imitation of protein-protein interactions. These polymers show nanomolar affinity for the target and neutralize their functions; thus, they are of significant interest as a cost-effective protein affinity reagent. We have been developing synthetic polymer nanoparticles (NPs) that bind to target peptides and proteins by the inclusion of several functional monomers, such as charged and hydrophobic monomers. In this review, the focus is on the design of synthetic polymer NPs that bind to target molecules for disease therapy. We succeeded in neutralization of toxic peptides and signaling proteins both in vitro and in vivo. Additionally, linear polymers were modified on a lipid nanoparticle surface to improve polymer biodistribution. Our recent findings should provide useful information for the development of abiotic protein affinity reagents.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/síntesis química , Diseño de Fármacos , Nanopartículas/administración & dosificación , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Animales , Humanos , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Neoplasias/metabolismo , Polímeros/administración & dosificación , Polímeros/síntesis química , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Regul Toxicol Pharmacol ; 120: 104857, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33387566

RESUMEN

Pharmaceutical and biotechnology companies rarely disclose their use of translational emerging safety biomarkers (ESBs) during drug development, and the impact of ESB use on the speed of drug development remains unclear. A cross-industry survey of 20 companies of varying size was conducted to understand current trends in ESB use and future use prospects. The objectives were to: (1) determine current ESB use in nonclinical and clinical drug development and impact on asset advancement; (2) identify opportunities, gaps, and challenges to greater ESB implementation; and (3) benchmark perspectives on regulatory acceptance. Although ESBs were employed in only 5-50% of studies/programs, most companies used ESBs to some extent, with larger companies demonstrating greater nonclinical use. Inclusion of ESBs in investigational new drug applications (INDs) was similar across all companies; however, differences in clinical trial usage could vary among the prevailing health authority (HA). Broader implementation of ESBs requires resource support, cross-industry partnerships, and collaboration with HAs. This includes generating sufficient foundational data, demonstrating nonclinical to clinical translatability and practical utility, and clearly written criteria by HAs to enable qualification. If achieved, ESBs will play a critical role in the development of next-generation, translationally-tailored standard laboratory tests for drug development.


Asunto(s)
Biomarcadores Farmacológicos/metabolismo , Ensayos Clínicos como Asunto/normas , Industria Farmacéutica/normas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Encuestas y Cuestionarios , Animales , Ensayos Clínicos como Asunto/métodos , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/normas , Industria Farmacéutica/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Predicción , Humanos , Preparaciones Farmacéuticas/metabolismo , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
18.
Nanomedicine ; 32: 102342, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33253922

RESUMEN

Acute kidney injury (AKI) is a life-threatening disease without effective treatment. The utilization of curcumin (Cur) for the treatment of AKI is still facing challenges due to its poor water-solubility and low bioavailability. Herein, kidney-targeted octenyl succinic anhydride-grafted fucoidan loaded with Cur (OSA-Fucoidan/Cur) was fabricated for synergistic treatment of AKI. It was found that OSA-Fucoidan/Cur micelles had a sustained drug release behavior and excellent physicochemical stability. Cellular uptake studies demonstrated that the specific binding between fucoidan and P-selectin overexpressed on H2O2-stimulated HUVECs contributed to the higher internalization of OSA-Fucoidan/Cur micelles by the cells. In addition, OSA-Fucoidan micelles exhibited an ideal kidney-targeted characteristic in lipopolysaccharide (LPS)-induced AKI mice. In vivo studies showed that the combination of Cur and OSA-Fucoidan endowed the OSA-Fucoidan/Cur micelles with synergistically anti-inflammatory and antioxidant abilities, thereby largely enhancing the therapeutic efficacy of AKI. Therefore, OSA-Fucoidan/Cur micelles may represent a potential kidney-targeted nanomedicine for effective treatment of AKI.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Portadores de Fármacos/química , Micelas , Selectina-P/antagonistas & inhibidores , Polisacáridos/química , Lesión Renal Aguda/patología , Animales , Antioxidantes/farmacología , Curcumina/farmacología , Curcumina/uso terapéutico , Liberación de Fármacos , Endocitosis/efectos de los fármacos , Semivida , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones Endogámicos ICR , Anhídridos Succínicos/química , Distribución Tisular/efectos de los fármacos
19.
Nanomedicine ; 32: 102315, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33065253

RESUMEN

Intracellular antibody delivery into live cells has significant implications for research and therapeutic applications. However, many delivery systems lack potency due to low uptake and/or endosomal entrapment and understanding of intracellular delivery processes is lacking. Herein, we studied the cellular uptake, intracellular trafficking and targeting of antibodies using our previously developed Hex antibody nanocarrier. We demonstrated Hex-antibodies were internalized through multiple endocytic routes into lysosomes and provide evidence of endo/lysosomal disruption and Hex-antibody release to the cytosol. Cytosolic antibodies retained their bioactivity for at least 24 h. Functional effect of Hex delivered anti-STAT3 antibodies was evidenced by inhibition of nuclear translocation of cytosolic transcription factor STAT3. This study has generated understanding of key steps in the Hex intracellular antibody delivery system and will facilitate the development of effective cytosolic antibody delivery and applications in both the therapeutic and research domains.


Asunto(s)
Anticuerpos/farmacología , Citosol/metabolismo , Sistemas de Liberación de Medicamentos , Espacio Intracelular/metabolismo , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Portadores de Fármacos/química , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Membrana Eritrocítica/efectos de los fármacos , Membrana Eritrocítica/metabolismo , Inmunoglobulina G/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Conejos , Distribución Tisular/efectos de los fármacos , Pavos
20.
Int J Mol Sci ; 22(22)2021 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-34830471

RESUMEN

Cardiovascular diseases (CVD), with myocardial infarction (MI) being one of the crucial components, wreak havoc in developed countries. Advanced imaging technologies are required to obtain quick and widely available diagnostic data. This paper describes a multimodal approach to in vivo perfusion imaging using the novel SYN1 tracer based on the fluorine-18 isotope. The NOD-SCID mice were injected intravenously with SYN1 or [18F] fluorodeoxyglucose ([18F]-FDG) radiotracers after induction of the MI. In all studies, the positron emission tomography-computed tomography (PET/CT) technique was used. To obtain hemodynamic data, mice were subjected to magnetic resonance imaging (MRI). Finally, the biodistribution of the SYN1 compound was performed using Wistar rat model. SYN1 showed normal accumulation in mouse and rat hearts, and MI hearts correctly indicated impaired cardiac segments when compared to [18F]-FDG uptake. In vivo PET/CT and MRI studies showed statistical convergence in terms of the size of the necrotic zone and cardiac function. This was further supported with RNAseq molecular analyses to correlate the candidate function genes' expression, with Serpinb1c, Tnc and Nupr1, with Trem2 and Aldolase B functional correlations showing statistical significance in both SYN1 and [18F]-FDG. Our manuscript presents a new fluorine-18-based perfusion radiotracer for PET/CT imaging that may have importance in clinical applications. Future research should focus on confirmation of the data elucidated here to prepare SYN1 for first-in-human trials.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Corazón/diagnóstico por imagen , Infarto del Miocardio/genética , Proteínas de Neoplasias/genética , Serpinas/genética , Tenascina/genética , Animales , Medios de Contraste/farmacología , Fluorodesoxiglucosa F18/farmacología , Fructosa-Bifosfato Aldolasa/genética , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Humanos , Imagen por Resonancia Magnética , Masculino , Glicoproteínas de Membrana/genética , Ratones , Infarto del Miocardio/patología , Tomografía Computarizada por Tomografía de Emisión de Positrones , Ratas , Receptores Inmunológicos/genética , Distribución Tisular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA