Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Med Phys ; 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38873842

RESUMEN

BACKGROUND: Atherosclerosis is a condition in which an adhesive substance called plaque accumulates over time inside the arteries. Plaque buildup results in the constriction of arteries, causing a shortage of blood supply to tissues and organs. Removing atherosclerotic plaques controls the development of acute ischemic stroke and heart diseases. It remains imperative for positive patient outcomes. PURPOSE: This study sought to develop a minimally invasive technique for removing arterial plaques by applying focused ultrasound (FUS) energy on the metal surface of a nitinol catheter wire to induce inertial cavitation. The induced cavitation can deplete plaque mechanically inside the arteries, leading towards improved recanalization of blood vessels. METHODS: The enhanced cavitation effect induced by combining FUS with a metal catheter was first verified by exposing agar phantom gels with or without a 0.9-mm diameter nitinol wire to an acoustic field produced by a 0.5-MHz FUS transducer. The phenomenon was further confirmed in pork belly fat samples with or without a 3-mm diameter nitinol catheter wire. Cavitation was monitored by detecting the peaks of emitted ultrasound signals from the samples using a passive cavitation detector (PCD). Cavitation threshold values were determined by observing the jump in the peak amplitude of signals received by the PCD when the applied FUS peak negative pressure (PNP) increased. To simulate arterial plaque removal, FUS with or without a catheter was used to remove tissues from pork belly fat samples and the lipid cores of human atherosclerotic plaque samples using 2500-cycle FUS bursts at 10% duty cycle and a burst repetition rate of 20 Hz. Treatment outcomes were quantified by subtracting the weight of samples before treatment from the weight of samples after treatment. All measurements were repeated 5 times (n = 5) unless otherwise indicated, and paired t-tests were used to compare the means of two groups. A p-value of <0.05 will be considered significant. RESULTS: Our results showed that with a nitinol wire, the cavitation threshold in agar phantoms was reduced to 2.6 MPa from 4.3 MPa PNP when there was no nitinol wire in the focal region of FUS. For pork belly fat samples, cavitation threshold values were 1.0 and 2.0 MPa PNP, with and without a catheter wire, respectively. Pork belly fat tissues and lipid cores of atherosclerotic plaques were depleted at the interface between a catheter and the samples at 2 and 4 MPa FUS PNP, respectively. The results showed that with a catheter wire in the focal region of a 3-min FUS treatment session, 24.7 and 25.6 mg of lipid tissues were removed from pork belly fat and human atherosclerotic samples, respectively. In contrast, the FUS-only group showed no reduction in sample weight. The differences between FUS-only and FUS-plus-catheter groups were statistically significant (p < 0.001 for the treatment on pork belly samples, and p < 0.01 for the treatment on human atherosclerotic samples). CONCLUSION: This study demonstrated the feasibility of catheter-assisted FUS therapy for removing atherosclerotic plaques.

2.
J Pharm Sci ; 112(3): 680-690, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36306862

RESUMEN

Liquid-liquid phase separation is a phenomenon within biology whereby proteins can separate into dense and more dilute phases with distinct properties. Three antibodies that undergo liquid-liquid phase separation were characterized in the protein-rich and protein-poor phases. In comparison to the protein-poor phase, the protein-rich phase demonstrates more blue-shift tryptophan emissions and red-shifted amide I absorbances. Large changes involving conformational isomerization around disulfide bonds were observed using Raman spectroscopy. Amide I and protein fluorescence differences between the phases persisted to temperatures above the critical temperature but ceased at the temperature at which aggregation occurred. In addition, large changes occurred in the structural organization of water molecules within the protein-rich phase for all three antibodies. It is hypothesized that as the proteins have the same chemical potential in both phases, the protein viscosity is higher in the protein-rich phase resulting in slowed diffusion dependent protein aggregation in this phase. For all three antibodies we performed accelerated stability studies and found that the protein-rich phase aggregated at the same rate or slower than the protein-poor phase.


Asunto(s)
Anticuerpos Monoclonales , Espectrometría Raman , Anticuerpos Monoclonales/química , Concentración de Iones de Hidrógeno , Temperatura
3.
Mol Pharm ; 19(11): 4357-4369, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36282296

RESUMEN

CpG oligodeoxynucleotides are toll-like receptor 9 agonists capable of inducing potent pro-inflammatory immune responses. Although CpG oligodeoxynucleotides have shown promising antitumor effects, their systemic activity can trigger immune-related toxicity, limiting therapeutic application. We previously identified glatiramer acetate (GA), a cationic polypeptide approved for the treatment of relapsing-remitting multiple sclerosis, as an intratumoral delivery agent capable of complexing with CpG, thereby pinning it to the injection site and limiting systemic exposure. Here, we investigated whether the combination of CpG or GA-CpG polyplexes and intraperitoneal anti-PD-1 therapy would result in synergistic efficacy in AT84 and CT26 murine syngeneic models of head and neck and colon cancers, respectively. In both AT84 and CT26 tumor models, intratumoral CpG or GA-CpG treatment similarly suppressed tumor growth, but the efficacy was not amplified with anti-PD-1. Nevertheless, combination treatment increased cytotoxic T cell, helper T cell, and natural killer cell infiltration into AT84 tumors. Surprisingly, the combination of intratumoral GA and intraperitoneal anti-PD-1 treatment resulted in elevated systemic GM-CSF and IL-2 cytokine levels and demonstrated synergistic antitumor effects in the CT26 mouse tumor model. Moreover, tumors that responded most significantly to anti-PD-1 plus GA treatment showed increased markers of infiltration of CD4+ T cells and natural killer cells. Combinations of intratumoral GA or GA-CpG polyplexes with anti-PD-1 treatment warrant further investigation as combination cancer immunotherapy strategies.


Asunto(s)
Inmunoterapia , Neoplasias , Ratones , Animales , Acetato de Glatiramer/uso terapéutico , Inmunoterapia/métodos , Oligodesoxirribonucleótidos , Adyuvantes Inmunológicos/uso terapéutico , Adyuvantes Inmunológicos/farmacología , Neoplasias/tratamiento farmacológico , Línea Celular Tumoral
4.
J Pharm Sci ; 111(3): 690-698, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34774918

RESUMEN

Lipid nanoparticles (LNPs) containing mRNA can deliver genetic material to cells for use as vaccines or protein replacement therapies. We characterized the effect of solution pH on cationic LNPs containing green fluorescent protein (EGFP) mRNA and their transfection efficiency. We compared the structural and colloidal properties of mRNA LNPs with LNPs not containing mRNA and mRNA free in solution. We used a combination of biophysical technique to build a picture of the structure of the lipids and mRNA across pH and temperature in the form of an empirical phase diagram (EPD). A combination of Fourier-transform infrared (FTIR) spectroscopy and differential scanning calorimetry was used to investigate lipid phase behavior. The mRNA-LNPs transition from an inverse hexagonal phase at pH values below the pKa of the cationic lipid to a lamellar phase above the pKa. At higher temperatures the mRNA-LNPs also transitioned from an inverse hexagonal phase to a lamellar phase indicating the inverse hexagonal phase is more thermodynamically favorable. Based on circular dichroism, the mRNA within the LNP has more A form structure at pH values below the lipid pKa than above it. Optical density, zeta potential and dynamic light scattering measurements were used to probe the colloidal stability of the mRNA-LNPs. The particles were larger and more prone to aggregation below the pKa. A stability study was performed to relate the biophysical characteristics to the storage of the particles in solution at 4 and 25 °C. mRNA-LNPs had the highest transfection efficiency and stability at pH values below the pKa. However, there was a trade-off between the stability and aggregation propensity since at very low pH the particles were most prone to aggregation. We performed kinetic experiments to show that the time scale of the pH-dependent phase behavior is slow (6 hour transition) and the transition from lamellar to inverse hexagonal phases is irreversible. This suggests that the lamellar phase is less stable and kinetically trapped. Our findings deepen our structural understanding of mRNA-LNPs and will aid the development of related formulations.


Asunto(s)
Lípidos , Nanopartículas , Cationes , Concentración de Iones de Hidrógeno , Lípidos/química , Liposomas , Nanopartículas/química , ARN Mensajero/química , ARN Mensajero/genética , ARN Interferente Pequeño/genética
5.
Photodiagnosis Photodyn Ther ; 37: 102706, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34954388

RESUMEN

PURPOSE: Conventional photosensitizers for photodynamic therapy (PDT) typically have wide tissue distribution and poor water solubility. A hyaluronic acid (HA) polymeric nanoparticle with specific lymphatic uptake and highly water solubility was developed to deliver pyropheophorbide-a (PPa) for locally advanced head and neck squamous cell carcinoma (HNSCC) treatment. METHODS AND RESULTS: PPa was chemically conjugated to the HA polymeric nanoparticle via an adipic acid dihydrazide (ADH) linker. The conjugates were injected subcutaneously in a region near the tumor. Near-infrared (NIR) imaging was used to monitor distribution, and diode laser was used to activate PPa. The singlet oxygen generation efficiency of PPa was not affected by conjugation to HA nanoparticles at a PPa loading degree of 1.89 w.t.%. HA-ADH-PPa inhibited human HNSCC MDA-1986 cell growth only when photo-irradiation was applied. After HA-ADH-PPa treatment and radiation, NU/NU mice bearing human HNSCC MDA-1986 tumors showed reduced tumor growth and significantly enhanced survival time compared with an untreated group (p < 0.05). CONCLUSIONS: These results demonstrate that HA-ADH-PPa could be useful for in vivo locoregional photodynamic therapy of HNSCC.


Asunto(s)
Neoplasias de Cabeza y Cuello , Nanopartículas , Fotoquimioterapia , Animales , Línea Celular Tumoral , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Ácido Hialurónico/farmacología , Ratones , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico
6.
Anal Biochem ; 634: 114425, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34678250

RESUMEN

Therapeutic proteins (TPs) are exposed to various immune cells like macrophages and neutrophils, especially after subcutaneous (SC) administration. It is well known that the immune cells can generate reactive oxygen species (ROS) and this may lead to oxidation of TPs. The oxidation can occur in the SC tissue after SC administration, during distribution to the immune organs like lymph nodes and spleen, and even in the blood circulation. The oxidation can lead to alteration of their pharmacokinetics and efficacy. Therefore, it is important to study the oxidation of TPs in the biological matrices using ultra-pressure chromatography-mass spectrometry. Rat growth hormone (rGH) was selected as a test protein due to its similarity with human growth hormone (hGH), which is widely used for treatment of growth hormone deficiency. In this manuscript, we have summarized sample processing strategy and ultra-pressure chromatography-mass spectrometry methodology to identify rGH and its degradation products after ex-vivo incubation with rat SC tissue, and in vitro incubation with rat splenocytes and canine peripheral blood mononuclear cells (cPBMCs) as a model foreign host species. We did not observe oxidation of rGH in these biological matrices. This could be due to very minor yields of oxidation products, lack of sensitivity of the mass spectrometry method, loss of protein during sample processing, rapid turnover of oxidized protein or a combination of all factors.


Asunto(s)
Hormona del Crecimiento/farmacología , Leucocitos Mononucleares/metabolismo , Tejido Subcutáneo/metabolismo , Animales , Cromatografía/métodos , Perros , Hormona del Crecimiento/administración & dosificación , Hormona del Crecimiento/farmacocinética , Hormona de Crecimiento Humana/farmacología , Humanos , Sistema Inmunológico/metabolismo , Inyecciones Subcutáneas , Masculino , Espectrometría de Masas/métodos , Oxidación-Reducción , Ratas , Especies Reactivas de Oxígeno/metabolismo , Bazo/metabolismo
7.
Med Phys ; 48(8): 4128-4138, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34214203

RESUMEN

PURPOSE: This study aimed to test the feasibility of combined ultrasound and laser technique, namely, ultrasound-assisted endovascular laser thrombolysis (USELT), for thrombolysis by conducting in vivo tests in a rabbit thrombosis model. MATERIALS AND METHODS: An acute thrombus was created in the right jugular vein of rabbit and then was treated with ultrasound only, laser only, and USELT to dissolve the blood clot. A total of 20 rabbits were used. Out of which, the first three rabbits were used to titrate the laser and ultrasound parameters. Then, five rabbits were treated with ultrasound only, five rabbits were treated with laser only, and seven rabbits were treated with USELT. During USELT, 532-nm laser pulses were delivered endovascularly directly to the clot through a fiber optic, and 0.5 MHz ultrasound pulses were applied noninvasively to the same region. A laser fluence of 4 to 12 mJ/cm2 and ultrasound amplitude of 1 to 2 MPa were used. Recanalization of the jugular vein was assessed by performing ultrasound Doppler imaging immediately after the treatment. The maximum blood flow speed after the treatment as compared to its value before the treatment was used to calculate the blood flow recovery in vessel. RESULTS: The blood flow was fully recovered (100%) in three rabbits, partially recovered in two rabbits (more than 50% and less than 100%) with mean percentage recovery of 69.73% and poorly recovered in two rabbits (<50%) with mean percentage recovery of 6.2% in the USELT group. In contrast, the treatment group with ultrasound or laser alone did not show recanalization of vein in any case, all the five rabbits were poorly/not recovered with a mean percentage recovery of 0%. CONCLUSIONS: The USELT technology was shown to effectively dissolve the blood clots in an acute rabbit jugular vein thrombosis model.


Asunto(s)
Trombosis , Animales , Estudios de Factibilidad , Rayos Láser , Conejos , Terapia Trombolítica , Trombosis/diagnóstico por imagen , Trombosis/terapia , Ultrasonografía
8.
J Control Release ; 336: 410-432, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34171445

RESUMEN

With the significant drawbacks of conventional cancer chemotherapeutics, cancer immunotherapy has demonstrated the ability to eradicate cancer cells and circumvent multidrug resistance (MDR) with fewer side effects than traditional cytotoxic therapies. Various immunotherapeutic agents have been investigated for that purpose including checkpoint inhibitors, cytokines, monoclonal antibodies and cancer vaccines. All these agents aid immune cells to recognize and engage tumor cells by acting on tumor-specific pathways, antigens or cellular targets. However, immunotherapeutics are still associated with some concerns such as off-target side effects and poor pharmacokinetics. Nanomedicine may resolve some limitations of current immunotherapeutics such as localizing delivery, controlling release and enhancing the pharmacokinetic profile. Herein, we discuss recent advances of immunotherapeutic agents with respect to their development and biological mechanisms of action, along with the advantages that nanomedicine strategies lend to immunotherapeutics by possibly improving therapeutic outcomes and minimizing side effects.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Biología , Humanos , Inmunoterapia , Nanomedicina , Neoplasias/terapia
9.
Med Phys ; 48(2): 579-586, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33280145

RESUMEN

PURPOSE: The combination of laser and ultrasound can significantly improve the efficiency of thrombolysis through an enhanced cavitation effect. We developed a fiber optics-based laser-ultrasound thrombolysis device and tested the feasibility and efficiency of this technology for restoring blood flow in an in vitro blood clot model. METHODS: An in vitro blood flow-clot model was setup, and then an endovascular laser thrombolysis system was combined with high-intensity focused ultrasound to remove the clot. The laser and ultrasound pulses were synchronized and delivered to the blood clot concurrently. The laser pulses of 532 nm were delivered to the blood clot endovascularly through an optical fiber, whereas the ultrasound pulses of 0.5 MHz were applied noninvasively to the same region. Effectiveness of thrombolysis was evaluated by the ability to restore blood flow, which was monitored by ultrasound Doppler. RESULTS: As laser powers increased, the ultrasound threshold pressures for effective thrombolysis decreased. For laser fluence levels of 0, 2, and 4 mJ/cm2 , the average negative ultrasound threshold pressures were 1.26 ± 0.114, 1.05 ± 0.181, and 0.59 ± 0.074 MPa, respectively. The periods of time needed to achieve effective thrombolysis were measured at 0.8, 2, and 4 mJ/cm2 laser fluence levels and 0.42, 0.70, and 0.98 MPa negative ultrasound pressures. In general, thrombolysis could be achieved more rapidly with higher laser powers or ultrasound pressures. CONCLUSIONS: Effective thrombolysis can be achieved by combining endovascular laser with noninvasive ultrasound at relatively low power and pressure levels, which can potentially improve both the treatment efficiency and safety.


Asunto(s)
Ultrasonido Enfocado de Alta Intensidad de Ablación , Trombosis , Humanos , Rayos Láser , Terapia Trombolítica , Ultrasonografía
10.
Macromol Biosci ; 20(12): e2000251, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32924274

RESUMEN

Designing an in vitro model of the tumor extracellular microenvironment to screen intratumoral drugs is an active challenge. As recent clinical successes of human intratumoral therapies stimulate research on intratumoral delivery, a need for a 3D tumor model to screen intratumoral therapies arises. When injecting the drug formulation directly into the tumor, the biophysics affecting intratumoral retention must be considered; especially for biologic therapies, which may be dominated by extracellular transport mechanisms. Fibrotic regions in solid tumors are typically rich in collagen I fibers. Using shear rheology, head and neck tumors with higher collagen density show a higher stiffness. Similarly, the stiffness of the hyaluronic acid (HA) hydrogel models is increased by adding collagen fibers to model the bulk biomechanical properties of solid tumors. HA hydrogels are then used as intratumoral injection site simulators to model in vitro the retention of glatiramer acetate (GA) and polyethylene glycol (PEG) administered intratumorally. Both compounds are also injected in murine tumors and retention is studied ex vivo for comparison. Retention of GA in the hydrogels is significantly longer than PEG, analogous to the solid tumors, suggesting the utility of HA hydrogels with collagen I fibers for screening extracellular drug transport after intratumoral administration.


Asunto(s)
Materiales Biocompatibles/farmacología , Sistemas de Liberación de Medicamentos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Hidrogeles/farmacología , Animales , Materiales Biocompatibles/metabolismo , Línea Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Composición de Medicamentos , Acetato de Glatiramer/química , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Ácido Hialurónico/química , Hidrogeles/química , Ratones , Polietilenglicoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Control Release ; 326: 203-221, 2020 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-32673633

RESUMEN

Cancer therapies aim to kill tumor cells directly or engage the immune system to fight malignancy. Checkpoint inhibitors, oncolytic viruses, cell-based immunotherapies, cytokines, and adjuvants have been applied to prompt the immune system to recognize and attack cancer cells. However, systemic exposure of cancer therapies can induce unwanted adverse events. Intratumoral administration of potent therapies utilizes small amounts of drugs, in an effort to minimize systemic exposure and off-target toxicities. Here, we discuss the properties of the tumor microenvironment and transport considerations for intratumoral drug delivery. Specifically, we consider various tumor tissue factors and physicochemical factors that can affect tumor retention after intratumoral injection. We also review approved and clinical-stage intratumoral therapies and consider how the molecular and biophysical properties (e.g. size and charge) of these therapies influences intratumoral transport (e.g. tumor retention and cellular uptake). Finally, we offer a critical review and highlight several emerging approaches to promote tumor retention and limit systemic exposure of potent intratumoral therapies.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Preparaciones Farmacéuticas , Humanos , Inmunoterapia , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
12.
J Pharm Sci ; 109(1): 191-205, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31408633

RESUMEN

The intradermal (ID) and subcutaneous (SC) routes are commonly used for therapeutic proteins (TPs) and vaccines; however, the bioavailability of TPs is typically less than small molecule drugs given via the same routes. Proteolytic enzymes in the dermal, SC, and lymphatic tissues may be responsible for the loss of TPs. In addition, the TPs may be exposed to reactive oxygen species generated in the SC tissue and the lymphatic system in response to injection-related trauma and impurities within the formulation. The reactive oxygen species can oxidize TPs to alter their efficacy and immunogenicity potential. Mechanistic understandings of the dominant proteolysis and oxidative routes are useful in the drug discovery process, formulation development, and to assess the potential for immunogenicity and altered pharmacokinetics (PK). Furthermore, in vitro tools representing the ID or SC and lymphatic system can be used to evaluate the extent of proteolysis of the TPs after the injection and before systemic entry. The in vitro clearance data may be included in physiologically based pharmacokinetic models for improved PK predictions. In this review, we have summarized various physiological factors responsible for proteolysis and oxidation of TPs after ID and SC administration.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/metabolismo , Glicoproteínas/administración & dosificación , Glicoproteínas/metabolismo , Proteolisis , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos , Inyecciones Intradérmicas/métodos , Inyecciones Subcutáneas/métodos , Sistema Linfático/efectos de los fármacos , Sistema Linfático/metabolismo , Oxidación-Reducción
13.
Bioorg Med Chem Lett ; 30(2): 126788, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31784317

RESUMEN

Small molecule agonists of TLR7/8, such as imidazoquinolines, are validated agonists for the treatment of cancer and for use in vaccine adjuvants. Imidazoquinolines have been extensively modified to understand the structure-activity relationship (SAR) at the N1- and C2-positions resulting in the clinical drug imiquimod, resiquimod, and several other highly potent analogues. However, the SAR of the aryl ring has not been fully elucidated in the literature. This initial study examines the SAR of C7-substituted imidazoquinolines. These compounds not only demonstrated that TLR7/8 tolerate changes at the C7-position but can increase potency and change their cytokine profiles. The most notable TLR7/8 agonists developed from this study 5, 8, and 14 which are up to 4-fold and 2-fold more active than resiquimod for TLR8 and/or TLR7, respectively, and up to 100-fold more active than the FDA approved imiquimod for TLR7.


Asunto(s)
Quinolinas/química , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Sitios de Unión , Citocinas/metabolismo , Humanos , Imidazoles/química , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Simulación del Acoplamiento Molecular , Quinolinas/metabolismo , Relación Estructura-Actividad , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo
14.
J Pharm Sci ; 108(10): 3372-3381, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31216451

RESUMEN

Oxidation of therapeutic proteins (TPs) can lead to changes in their pharmacokinetics, biological activity and immunogenicity. Metal impurities such as iron are known to increase oxidation of TPs, but nanoparticulate metals have unique physical and chemical properties compared to the bulk material or free metal ions. Iron oxide nanoparticles (IONPs) may originate from equipment used in the manufacturing of TPs or from needles during injection. In this study, the impact of IONPs on oxidation of a model protein, rat growth hormone (rGH), was investigated under chemical stress. Hydrogen peroxide (H2O2)- and 2,2'-azobis (2-methylpropionamidine) dihydrochloride oxidized methionine residues of rGH, but unexpectedly, oxidation was suppressed in the presence of IONPs compared to a phosphate buffer control. Fourier transform infrared spectroscopy indicated splitting of the α-helical absorbance band in the presence of IONPs, whereas circular dichroism spectra showed a reduced α-helical contribution with increasing temperature for both rGH and rGH-IONP mixtures. The results collectively indicate that IONPs can increase the chemical stability of rGH by altering the kinetics and preference of amino acid residues that are oxidized, although the changes in protein secondary structure by IONPs may lead to alterations of physical stability.


Asunto(s)
Compuestos Férricos/química , Hormona del Crecimiento/química , Hierro/química , Nanopartículas/química , Oxidación-Reducción/efectos de los fármacos , Estructura Secundaria de Proteína/efectos de los fármacos , Aminoácidos/química , Animales , Dicroismo Circular , Peróxido de Hidrógeno/química , Conformación Proteica en Hélice alfa/efectos de los fármacos , Ratas
15.
J Control Release ; 306: 165-176, 2019 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-31173789

RESUMEN

The toll-like receptor 7 and 8 (TLR7/8) agonist Resiquimod (R848) has been recognized as a promising immunostimulator for the treatment of cutaneous cancers in multiple clinical trials. However, systemic administration of R848 often results in strong immune-related toxicities while having limited therapeutic effects to the tumor. In the present study, a prodrug-based nanocarrier delivery system was developed that exhibited high therapeutic efficiency. R848 was conjugated to α-tocopherol to constitute an R848-Toco prodrug, followed by formulating with a tocopherol-modified hyaluronic acid (HA-Toco) as a polymeric nano-suspension. In vitro evaluation showed that the delivery system prolonged the release kinetics while maintaining TLR agonist activities. When administered subcutaneously, the nano-suspension formed a depot at the injection site, inducing localized immune responses without systemic expansion. This formulation also suppressed tumor growth and recruited immune cells to the tumor in a murine model of head and neck cancer. In a preclinical canine study of spontaneous mast cell tumors, the treatment led to a 67% response rate (three partial remissions and one complete remission).


Asunto(s)
Antineoplásicos/farmacología , Imidazoles/administración & dosificación , Factores Inmunológicos/farmacología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Animales , Citocinas/metabolismo , Perros , Composición de Medicamentos , Evaluación Preclínica de Medicamentos , Liberación de Fármacos , Humanos , Imidazoles/química , Imidazoles/farmacología , Ratones , Ratones Endogámicos C3H , Conejos , Suspensiones
16.
J Control Release ; 293: 36-47, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30414463

RESUMEN

Glatiramer acetate (GA) is widely prescribed for the treatment of relapsing-remitting multiple sclerosis, however, the mechanism of action is still not fully understood. We investigated the structural properties of GA and examined alterations to the drug upon injection into the subcutaneous space. First, a variety of biophysical characterization techniques were employed to characterize GA in solution. GA was found to exist as alpha helices in solution with a hydrodynamic radius of ~3 nm in size. To simulate GA behavior at the site of injection, GA was injected into a solution of 1.5 MDa hyaluronic acid (HA). Visible aggregates were observed immediately upon injection and subsequent testing indicated aggregation was driven by electrostatic interactions between the positively-charged GA and negatively-charged HA. In vivo testing confirmed GA formed spherical particles in the nano- to micrometer size range, suggesting this mechanism contributes to persistence at the injection site and in draining lymph nodes. The aggregates were found to associate with glycosaminoglycans, suggesting an electrostatic mechanism of induced aggregation like the simulated injection. These novel observations may help explain the complex immunomodulatory mechanisms of GA and adverse injection site reactions seen in patients.


Asunto(s)
Acetato de Glatiramer , Inmunosupresores , Animales , Femenino , Acetato de Glatiramer/administración & dosificación , Acetato de Glatiramer/química , Acetato de Glatiramer/farmacocinética , Ácido Hialurónico/química , Inmunosupresores/administración & dosificación , Inmunosupresores/química , Inmunosupresores/farmacocinética , Inyecciones Subcutáneas , Ganglios Linfáticos/metabolismo , Ratones , Músculo Esquelético/metabolismo , Nanopartículas , Electricidad Estática
17.
Drug Deliv Transl Res ; 8(5): 1592, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30073575

RESUMEN

In the XML of the original article, M. Laird Forrest's name was tagged incorrectly. M. Laird is his first name.

18.
Pharm Res ; 35(9): 180, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30046910

RESUMEN

One of the authors has his name incorrectly indexed in PubMed and SpringerLink as "Laird Forrest M" (last name "Laird Forrest"). His name should index as "Forrest M. Laird" with last name as "Forrest".

19.
Pharm Res ; 35(8): 162, 2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29931580

RESUMEN

PURPOSE: To investigate influence of inflammation on metabolism and pharmacokinetics (PK) of midazolam (MDZ) and construct a semi-physiologically based pharmacokinetic (PBPK) model to predict PK in mice with inflammatory disease. METHODS: Glucose-6-phosphate isomerase (GPI)-mediated inflammation was used as a preclinical model of arthritis in DBA/1 mice. CYP3A substrate MDZ was selected to study changes in metabolism and PK during the inflammation. The semi-PBPK model was constructed using mouse physiological parameters, liver microsome metabolism, and healthy animal PK data. In addition, serum cytokine, and liver-CYP (cytochrome P450 enzymes) mRNA levels were examined. RESULTS: The in vitro metabolite formation rate was suppressed in liver microsomes prepared from the GPI-treated mice as compared to the healthy mice. Further, clearance of MDZ was reduced during inflammation as compared to the healthy group. Finally, the semi-PBPK model was used to predict PK of MDZ after GPI-mediated inflammation. IL-6 and TNF-α levels were elevated and liver-cyp3a11 mRNA was reduced after GPI treatment. CONCLUSION: The semi-PBPK model successfully predicted PK parameters of MDZ in the disease state. The model may be applied to predict PK of other drugs under disease conditions using healthy animal PK and liver microsomal data as inputs.


Asunto(s)
Adyuvantes Anestésicos/farmacocinética , Inflamación/metabolismo , Midazolam/farmacocinética , Adyuvantes Anestésicos/metabolismo , Animales , Citocromo P-450 CYP3A/metabolismo , Glucosa-6-Fosfato Isomerasa/metabolismo , Humanos , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Midazolam/metabolismo , Modelos Biológicos
20.
Mol Cancer Res ; 16(6): 961-973, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29545475

RESUMEN

FOXM1 transcription factor network is activated in over 84% of cases in high-grade serous ovarian cancer (HGSOC), and FOXM1 upregulates the expression of genes involved in the homologous recombination (HR) DNA damage and repair (DDR) pathway. However, the role of FOXM1 in PARP inhibitor response has not yet been studied. This study demonstrates that PARP inhibitor (PARPi), olaparib, induces the expression and nuclear localization of FOXM1. On the basis of ChIP-qPCR, olaparib enhances the binding of FOXM1 to genes involved in HR repair. FOXM1 knockdown by RNAi or inhibition by thiostrepton decreases FOXM1 expression, decreases the expression of HR repair genes, such as BRCA1 and RAD51, and enhances sensitivity to olaparib. Comet and PARP trapping assays revealed increases in DNA damage and PARP trapping in FOXM1-inhibited cells treated with olaparib. Finally, thiostrepton decreases the expression of BRCA1 in rucaparib-resistant cells and enhances sensitivity to rucaparib. Collectively, these results identify that FOXM1 plays an important role in the adaptive response induced by olaparib and FOXM1 inhibition by thiostrepton induces "BRCAness" and enhances sensitivity to PARP inhibitors.Implications: FOXM1 inhibition represents an effective strategy to overcome resistance to PARPi, and targeting FOXM1-mediated adaptive pathways may produce better therapeutic effects for PARP inhibitors. Mol Cancer Res; 16(6); 961-73. ©2018 AACR.


Asunto(s)
Antineoplásicos/efectos adversos , Proteína Forkhead Box M1/genética , Ftalazinas/efectos adversos , Piperazinas/efectos adversos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Ftalazinas/farmacología , Piperazinas/farmacología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...