Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Nat Mater ; 19(8): 910-920, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32341511

RESUMEN

Long-acting cabotegravir (CAB) extends antiretroviral drug administration from daily to monthly. However, dosing volumes, injection site reactions and health-care oversight are obstacles towards a broad usage. The creation of poloxamer-coated hydrophobic and lipophilic CAB prodrugs with controlled hydrolysis and tissue penetrance can overcome these obstacles. To such ends, fatty acid ester CAB nanocrystal prodrugs with 14, 18 and 22 added carbon chains were encased in biocompatible surfactants named NMCAB, NM2CAB and NM3CAB and tested for drug release, activation, cytotoxicity, antiretroviral activities, pharmacokinetics and biodistribution. Pharmacokinetics studies, performed in mice and rhesus macaques, with the lead 18-carbon ester chain NM2CAB, showed plasma CAB levels above the protein-adjusted 90% inhibitory concentration for up to a year. NM2CAB, compared with NMCAB and NM3CAB, demonstrated a prolonged drug release, plasma circulation time and tissue drug concentrations after a single 45 mg per kg body weight intramuscular injection. These prodrug modifications could substantially improve CAB's effectiveness.


Asunto(s)
Antirretrovirales/metabolismo , Nanoestructuras/química , Profármacos/química , Profármacos/metabolismo , Piridonas/metabolismo , Animales , Antirretrovirales/farmacología , Antirretrovirales/toxicidad , Transporte Biológico , Preparaciones de Acción Retardada , Composición de Medicamentos , Interacciones Farmacológicas , Estabilidad de Medicamentos , Ratones , Piridonas/farmacología , Piridonas/toxicidad
2.
J Pharmacol Exp Ther ; 365(2): 272-280, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29476044

RESUMEN

Antiretroviral drug (ARV) metabolism is linked largely to hepatic cytochrome P450 activity. One ARV drug class known to be metabolized by intestinal and hepatic CYP3A are the protease inhibitors (PIs). Plasma drug concentrations are boosted by CYP3A inhibitors such as cobisistat and ritonavir (RTV). Studies of such drug-drug interactions are limited since the enzyme pathways are human specific. While immune-deficient mice reconstituted with human cells are an excellent model to study ARVs during human immunodeficiency virus type 1 (HIV-1) infection, they cannot reflect human drug metabolism. Thus, we created a mouse strain with the human pregnane X receptor, constitutive androstane receptor, and CYP3A4/7 genes on a NOD.Cg-Prkdcscid Il2rgtm1Sug /JicTac background (hCYP3A-NOG) and used them to evaluate the impact of human CYP3A metabolism on ARV pharmacokinetics. In proof-of-concept studies we used nanoformulated atazanavir (nanoATV) with or without RTV. NOG and hCYP3A-NOG mice were treated weekly with 50 mg/kg nanoATV alone or boosted with nanoformulated ritonavir (nanoATV/r). Plasma was collected weekly and liver was collected at 28 days post-treatment. Plasma and liver atazanavir (ATV) concentrations in nanoATV/r-treated hCYP3A-NOG mice were 2- to 4-fold higher than in replicate NOG mice. RTV enhanced plasma and liver ATV concentrations 3-fold in hCYP3A-NOG mice and 1.7-fold in NOG mice. The results indicate that human CYP3A-mediated drug metabolism is reduced compared with mouse and that RTV differentially affects human gene activity. These differences can affect responses to PIs in humanized mouse models of HIV-1 infection. Importantly, hCYP3A-NOG mice reconstituted with human immune cells can be used for bench-to-bedside translation.


Asunto(s)
Fármacos Anti-VIH/farmacología , Citocromo P-450 CYP3A/genética , Receptor X de Pregnano/genética , Receptores Citoplasmáticos y Nucleares/genética , Animales , Fármacos Anti-VIH/farmacocinética , Receptor de Androstano Constitutivo , Interacciones Farmacológicas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Distribución Tisular , Investigación Biomédica Traslacional
3.
Nanomedicine ; 12(1): 109-22, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26472049

RESUMEN

During studies to extend the half-life of crystalline nanoformulated antiretroviral therapy (nanoART) the mixed lineage kinase-3 inhibitor URMC-099, developed as an adjunctive neuroprotective agent was shown to facilitate antiviral responses. Long-acting ritonavir-boosted atazanavir (nanoATV/r) nanoformulations co-administered with URMC-099 reduced viral load and the numbers of HIV-1 infected CD4+ T-cells in lymphoid tissues more than either drug alone in infected humanized NOD/SCID/IL2Rγc-/- mice. The drug effects were associated with sustained ART depots. Proteomics analyses demonstrated that the antiretroviral responses were linked to affected phagolysosomal storage pathways leading to sequestration of nanoATV/r in Rab-associated recycling and late endosomes; sites associated with viral maturation. URMC-099 administered with nanoATV induced a dose-dependent reduction in HIV-1p24 and reverse transcriptase activity. This drug combination offers a unique chemical marriage for cell-based viral clearance. From the Clinical Editor: Although successful in combating HIV-1 infection, the next improvement in antiretroviral therapy (nanoART) would be to devise long acting therapy, such as intra-cellular depots. In this report, the authors described the use of nanoformulated antiretroviral therapy given together with the mixed lineage kinase-3 inhibitor URMC-099, and showed that this combination not only prolonged drug half-life, but also had better efficacy. The findings are hoped to be translated into the clinical setting in the future.


Asunto(s)
Sulfato de Atazanavir/administración & dosificación , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , Nanocápsulas/química , Piridinas/administración & dosificación , Pirroles/administración & dosificación , Animales , Antirretrovirales/administración & dosificación , Terapia Antirretroviral Altamente Activa/métodos , Quimioterapia Combinada/métodos , Infecciones por VIH/diagnóstico , Humanos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Ratones , Ratones SCID , Nanocápsulas/administración & dosificación , Nanocápsulas/ultraestructura , Inhibidores de Proteínas Quinasas/administración & dosificación , Resultado del Tratamiento , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
4.
J Virol ; 88(17): 9504-13, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24920821

RESUMEN

UNLABELLED: Limitations of antiretroviral therapy (ART) include poor patient adherence, drug toxicities, viral resistance, and failure to penetrate viral reservoirs. Recent developments in nanoformulated ART (nanoART) could overcome such limitations. To this end, we now report a novel effect of nanoART that facilitates drug depots within intracellular compartments at or adjacent to the sites of the viral replication cycle. Poloxamer 407-coated nanocrystals containing the protease inhibitor atazanavir (ATV) were prepared by high-pressure homogenization. These drug particles readily accumulated in human monocyte-derived macrophages (MDM). NanoATV concentrations were ∼1,000 times higher in cells than those that could be achieved by the native drug. ATV particles in late and recycling endosome compartments were seen following pulldown by immunoaffinity chromatography with Rab-specific antibodies conjugated to magnetic beads. Confocal microscopy provided cross validation by immunofluorescent staining of the compartments. Mathematical modeling validated drug-endosomal interactions. Measures of reverse transcriptase activity and HIV-1 p24 levels in culture media and cells showed that such endosomal drug concentrations enhanced antiviral responses up to 1,000-fold. We conclude that late and recycling endosomes can serve as depots for nanoATV. The colocalization of nanoATV at endosomal sites of viral assembly and its slow release sped antiretroviral activities. Long-acting nanoART can serve as a drug carrier in both cells and subcellular compartments and, as such, can facilitate viral clearance. IMPORTANCE: The need for long-acting ART is significant and highlighted by limitations in drug access, toxicity, adherence, and reservoir penetrance. We propose that targeting nanoformulated drugs to infected tissues, cells, and subcellular sites of viral replication may improve clinical outcomes. Endosomes are sites for human immunodeficiency virus assembly, and increasing ART concentrations in such sites enhances viral clearance. The current work uncovers a new mechanism by which nanoART can enhance viral clearance over native drug formulations.


Asunto(s)
Antirretrovirales/farmacocinética , Endosomas/metabolismo , VIH-1/efectos de los fármacos , Macrófagos/metabolismo , Nanopartículas , Oligopéptidos/farmacocinética , Poloxámero/farmacocinética , Piridinas/farmacocinética , Antirretrovirales/farmacología , Sulfato de Atazanavir , Transporte Biológico , Células Cultivadas , Proteína p24 del Núcleo del VIH/análisis , VIH-1/crecimiento & desarrollo , Humanos , Microscopía Confocal , Microscopía Fluorescente , Modelos Teóricos , Oligopéptidos/farmacología , Poloxámero/farmacología , Piridinas/farmacología , Cultivo de Virus
5.
Nanomedicine ; 11(3): 715-29, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25652894

RESUMEN

Nanoparticulate delivery systems represent an area of particular promise for nanoneuromedicines. They possess significant potential for desperately needed therapies designed to combat a range of disorders associated with aging. As such, the field was selected as the focus for the 2014 meeting of the American Society for Nanomedicine. Regenerative, protective, immune modulatory, anti-microbial and anti-inflammatory products, or imaging agents are readily encapsulated in or conjugated to nanoparticles and as such facilitate the delivery of drug payloads to specific action sites across the blood-brain barrier. Diagnostic imaging serves to precisely monitor disease onset and progression while neural stem cell replacement can regenerate damaged tissue through control of stem cell fates. These, taken together, can improve disease burden and limit systemic toxicities. Such enabling technologies serve to protect the nervous system against a broad range of degenerative, traumatic, metabolic, infectious and immune disorders. From the clinical editor: Nanoneuromedicine is a branch of nanomedicine that specifically looks at the nervous system. In the clinical setting, a fundamental hurdle in nervous system disorders is due to an inherent inability of nerve cells to regenerate after damage. Nanotechnology can offer new approaches to overcome these challenges. This review describes recent developments in nanomedicine delivery systems that would affect stem cell repair and regeneration in the nervous system.


Asunto(s)
Envejecimiento , Sistemas de Liberación de Medicamentos/métodos , Nanomedicina/métodos , Nanoestructuras/uso terapéutico , Enfermedades del Sistema Nervioso/terapia , Células-Madre Neurales , Enfermedades del Sistema Nervioso/metabolismo
6.
J Proteome Res ; 12(5): 2282-94, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23544708

RESUMEN

Our laboratory developed long-acting nanoformulations of antiretroviral therapy (nanoART) to improve drug compliance, reduce toxicities, and facilitate access of drug to viral reservoirs. These all function to inevitably improve treatment of human immunodeficiency virus (HIV) infection. Formulations are designed to harness the carrying capacities of mononuclear phagocytes (MP; monocytes and macrophages) and to use these cells as Trojan horses for drug delivery. Such a drug distribution system limits ART metabolism and excretion while facilitating access to viral reservoirs. Our prior works demonstrated a high degree of nanoART sequestration in macrophage recycling endosomes with broad and sustained drug tissue biodistribution and depots with limited untoward systemic toxicities. Despite such benefits, the effects of particle carriage on the cells' functional capacities remained poorly understood. Thus, we employed pulsed stable isotope labeling of amino acids in cell culture to elucidate the macrophage proteome and assess any alterations in cellular functions that would affect cell-drug carriage and release kinetics. NanoART-MP interactions resulted in the induction of a broad range of activation-related proteins that can enhance phagocytosis, secretory functions, and cell migration. Notably, we now demonstrate that particle-cell interactions serve to enhance drug loading while facilitating drug tissue depots and transportation.


Asunto(s)
Fármacos Anti-VIH/farmacología , Portadores de Fármacos/farmacología , Macrófagos/metabolismo , Proteoma/metabolismo , Alquinos , Fármacos Anti-VIH/química , Sulfato de Atazanavir , Benzoxazinas/química , Benzoxazinas/farmacología , Movimiento Celular , Células Cultivadas , Quimiocinas/metabolismo , Ciclopropanos , Proteínas del Citoesqueleto/metabolismo , Portadores de Fármacos/química , Composición de Medicamentos , Humanos , Macrófagos/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Nanopartículas/química , Oligopéptidos/química , Oligopéptidos/farmacología , Tamaño de la Partícula , Fagocitosis/efectos de los fármacos , Canales de Potasio/metabolismo , Piridinas/química , Piridinas/farmacología , Ritonavir/química , Ritonavir/farmacología
7.
Nat Commun ; 13(1): 3226, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35680875

RESUMEN

Ultra-long-acting integrase strand transfer inhibitors were created by screening a library of monomeric and dimeric dolutegravir (DTG) prodrug nanoformulations. This led to an 18-carbon chain modified ester prodrug nanocrystal (coined NM2DTG) with the potential to sustain yearly dosing. Here, we show that the physiochemical and pharmacokinetic (PK) formulation properties facilitate slow drug release from tissue macrophage depot stores at the muscle injection site and adjacent lymphoid tissues following single parenteral injection. Significant plasma drug levels are recorded up to a year following injection. Tissue sites for prodrug hydrolysis are dependent on nanocrystal dissolution and prodrug release, drug-depot volume, perfusion, and cell-tissue pH. Each affect an extended NM2DTG apparent half-life recorded by PK parameters. The NM2DTG product can impact therapeutic adherence, tolerability, and access of a widely used integrase inhibitor in both resource limited and rich settings to reduce HIV-1 transmission and achieve optimal treatment outcomes.


Asunto(s)
Infecciones por VIH , Inhibidores de Integrasa VIH , Profármacos , Infecciones por VIH/tratamiento farmacológico , Compuestos Heterocíclicos con 3 Anillos , Humanos , Oxazinas/uso terapéutico , Piperazinas , Profármacos/farmacología , Piridonas/uso terapéutico
8.
J Immunol ; 183(1): 661-9, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19535632

RESUMEN

Antiretroviral therapy (ART) shows variable blood-brain barrier penetration. This may affect the development of neurological complications of HIV infection. In attempts to attenuate viral growth for the nervous system, cell-based nanoformulations were developed with the focus on improving drug pharmacokinetics. We reasoned that ART carriage could be facilitated within blood-borne macrophages traveling across the blood-brain barrier. To test this idea, an HIV-1 encephalitis (HIVE) rodent model was used where HIV-1-infected human monocyte-derived macrophages were stereotactically injected into the subcortex of severe combined immunodeficient mice. ART was prepared using indinavir (IDV) nanoparticles (NP, nanoART) loaded into murine bone marrow macrophages (BMM, IDV-NP-BMM) after ex vivo cultivation. IDV-NP-BMM was administered i.v. to mice resulting in continuous IDV release for 14 days. Rhodamine-labeled IDV-NP was readily observed in areas of HIVE and specifically in brain subregions with active astrogliosis, microgliosis, and neuronal loss. IDV-NP-BMM treatment led to robust IDV levels and reduced HIV-1 replication in HIVE brain regions. We conclude that nanoART targeting to diseased brain through macrophage carriage is possible and can be considered in developmental therapeutics for HIV-associated neurological disease.


Asunto(s)
Encéfalo/virología , Encefalitis Viral/tratamiento farmacológico , VIH-1/efectos de los fármacos , Indinavir/administración & dosificación , Macrófagos/trasplante , Macrófagos/virología , Nanocápsulas/administración & dosificación , Inmunodeficiencia Combinada Grave/tratamiento farmacológico , Animales , Disponibilidad Biológica , Células de la Médula Ósea/patología , Células de la Médula Ósea/virología , Encéfalo/patología , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Esquema de Medicación , Encefalitis Viral/metabolismo , VIH-1/crecimiento & desarrollo , Humanos , Indinavir/farmacocinética , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Inmunodeficiencia Combinada Grave/virología , Replicación Viral/efectos de los fármacos
9.
Mol Neurobiol ; 58(11): 5703-5721, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34390469

RESUMEN

Dolutegravir (DTG) is a first-line antiretroviral drug (ARV) used in combination therapy for the treatment of human immunodeficiency virus type-1 (HIV-1) infection. The drug is effective, safe, and well tolerated. Nonetheless, concerns have recently emerged for its usage in pregnant women or those of child-bearing age. Notably, DTG-based ARV regimens have been linked to birth defects seen as a consequence of periconceptional usages. To this end, uncovering an underlying mechanism for DTG-associated adverse fetal development outcomes has gained clinical and basic research interest. We now report that DTG inhibits matrix metalloproteinases (MMPs) activities that could affect fetal neurodevelopment. DTG is a broad-spectrum MMPs inhibitor and binds to Zn++ at the enzyme's catalytic domain. Studies performed in pregnant mice show that DTG readily reaches the fetal central nervous system during gestation and inhibits MMP activity. Postnatal screenings of brain health in mice pups identified neuroinflammation and neuronal impairment. These abnormalities persist as a consequence of in utero DTG exposure. We conclude that DTG inhibition of MMPs activities during gestation has the potential to affect prenatal and postnatal neurodevelopment.


Asunto(s)
Antirretrovirales/toxicidad , Compuestos Heterocíclicos con 3 Anillos/toxicidad , Inhibidores de la Metaloproteinasa de la Matriz/toxicidad , Defectos del Tubo Neural/inducido químicamente , Trastornos del Neurodesarrollo/inducido químicamente , Enfermedades Neuroinflamatorias/inducido químicamente , Oxazinas/toxicidad , Piperazinas/toxicidad , Piridonas/toxicidad , Animales , Antirretrovirales/farmacocinética , Antirretrovirales/farmacología , Encéfalo/embriología , Encéfalo/enzimología , Dominio Catalítico/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Compuestos Heterocíclicos con 3 Anillos/farmacocinética , Compuestos Heterocíclicos con 3 Anillos/farmacología , Masculino , Inhibidores de la Metaloproteinasa de la Matriz/farmacocinética , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Ratones Endogámicos C3H , Simulación del Acoplamiento Molecular , Defectos del Tubo Neural/embriología , Neuroimagen , Enfermedades Neuroinflamatorias/embriología , Oxazinas/farmacocinética , Oxazinas/farmacología , Piperazinas/farmacocinética , Piperazinas/farmacología , Placenta/química , Embarazo , Piridonas/farmacocinética , Piridonas/farmacología , Distribución Tisular , Zinc/metabolismo
10.
Antiviral Res ; 187: 104975, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33450312

RESUMEN

While muscle fatigue, pain and weakness are common co-morbidities in HIV-1 infected people, their underlying cause remain poorly defined. To this end, we evaluated whether the common antiretroviral drugs efavirenz (EFV), atazanavir (ATV) and ritonavir (RTV) could be a contributing factor by pertubating sarcoplasmic reticulum (SR) Ca2+ cycling. In live-cell imaging, EFV (6.0 µM), ATV (6.0 µM), and RTV (3.0 µM) elicited Ca2+ transients and blebbing of the plasma membranes of C2C12 skeletal muscle myotubes. Pretreating C2C12 skeletal muscle myotubes with the SR Ca2+ release channel blocker ryanodine (50 µM), slowed the rate and amplitude of Ca2+ release from and reuptake of Ca2+ into the SR. EFV, ATV and RTV (1 nM - 20 µM) potentiated and then displaced [3H] ryanodine binding to rabbit skeletal muscle ryanodine receptor Ca2+ release channel (RyR1). These drugs at concentrations 0.25-31.2 µM also increased and or decreased the open probability of RyR1 by altering its gating and conductance. ATV (≤5 µM) potentiated and >5µM inhibited the ability of sarco (endo)plasmic reticulum Ca2+-ATPase (SERCA1) to hydrolyze ATP and transport Ca2+. RTV (2.5-31.5 µM) dose-dependently inhibited SERCA1-mediated, ATP-dependent Ca2+ transport. EFV (0.25-31.5 µM) had no measurable effect on SERCA1's ability to hydrolyze ATP and transport Ca2+. These data support the notion that EFV, ATV and RTV could be contributing to skeletal muscle co-morbidities in PLWH by modulating SR Ca2+ homeostasis.


Asunto(s)
Fármacos Anti-VIH/efectos adversos , Calcio/metabolismo , Músculo Esquelético/efectos de los fármacos , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/efectos de los fármacos , Alquinos/efectos adversos , Animales , Sulfato de Atazanavir/efectos adversos , Benzoxazinas/efectos adversos , Línea Celular , Ciclopropanos/efectos adversos , Homeostasis , Ratones , Mioblastos/efectos de los fármacos , Conejos , Ritonavir/efectos adversos , Rianodina/farmacología , Retículo Sarcoplasmático/metabolismo , Imagen de Lapso de Tiempo
11.
Nat Commun ; 12(1): 3453, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34103484

RESUMEN

A once every eight-week cabotegravir (CAB) long-acting parenteral is more effective than daily oral emtricitabine and tenofovir disoproxil fumarate in preventing human immunodeficiency virus type one (HIV-1) transmission. Extending CAB dosing to a yearly injectable advances efforts for the elimination of viral transmission. Here we report rigor, reproducibility and mechanistic insights for a year-long CAB injectable. Pharmacokinetic (PK) profiles of this nanoformulated CAB prodrug (NM2CAB) are affirmed at three independent research laboratories. PK profiles in mice and rats show plasma CAB levels at or above the protein-adjusted 90% inhibitory concentration for a year after a single dose. Sustained native and prodrug concentrations are at the muscle injection site and in lymphoid tissues. The results parallel NM2CAB uptake and retention in human macrophages. NM2CAB nanocrystals are stable in blood and tissue homogenates. The long apparent drug half-life follows pH-dependent prodrug hydrolysis upon slow prodrug nanocrystal dissolution and absorption. In contrast, solubilized prodrug is hydrolyzed in hours in plasma and tissues from multiple mammalian species. No toxicities are observed in animals. These results affirm the pharmacological properties and extended apparent half-life for a nanoformulated CAB prodrug. The report serves to support the mechanistic design for drug formulation safety, rigor and reproducibility.


Asunto(s)
Liberación de Fármacos , Lípidos/química , Nanopartículas/química , Profármacos/farmacología , Piridonas/farmacocinética , Animales , Composición de Medicamentos , Endocitosis , Humanos , Cinética , Masculino , Ratones Endogámicos BALB C , Piridonas/administración & dosificación , Piridonas/sangre , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Distribución Tisular
12.
Expert Opin Drug Deliv ; 17(9): 1227-1238, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32552187

RESUMEN

INTRODUCTION: Despite significant advances in treatment and prevention of HIV-1 infection, poor adherence to daily combination antiretroviral therapy (ART) regimens remains a major obstacle toward achieving sustained viral suppression and prevention. Adherence to ART could also be compromised by adverse drug reactions and societal factors that limit access to therapy. Therefore, medicines that aim to improve adherence by limiting ART side effects, frequency of dosing and socially acceptable regimens are becoming more attractive. AREAS COVERED: This review highlights recent advances and challenges in the development of long-acting drug delivery strategies for HIV prevention and treatment. Approaches for extended oral and transdermal deliveries, microbicides, broadly neutralizing antibodies, and long-acting implantable and injectable deliveries are reviewed. EXPERT OPINION: Emerging approaches on long-acting antiretroviral therapies and broadly neutralizing antibody technologies are currently at various stages of development. Such efforts, if successful and become broadly accepted by clinicians and users, will provide newer and simpler options for prevention and treatment of HIV infection.


Asunto(s)
Fármacos Anti-VIH/administración & dosificación , Sistemas de Liberación de Medicamentos , Infecciones por VIH/tratamiento farmacológico , Administración Cutánea , Humanos
13.
Int J Nanomedicine ; 14: 6231-6247, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31496683

RESUMEN

PURPOSE: A palmitoylated prodrug of emtricitabine (FTC) was synthesized to extend the drug's half-life, antiretroviral activities and biodistribution. METHODS: A modified FTC prodrug (MFTC) was synthesized by palmitoyl chloride esterification. MFTC's chemical structure was evaluated by nuclear magnetic resonance. The created hydrophobic prodrug nanocrystals were encased into a poloxamer surfactant and the pharmacokinetics (PK), biodistribution and antiretroviral activities of the nanoformulation (NMFTC) were assessed. The conversion of MFTC to FTC triphosphates was evaluated. RESULTS: MFTC coated with poloxamer formed stable nanocrystals (NMFTC). NMFTC demonstrated an average particle size, polydispersity index and zeta potential of 350 nm, 0.24 and -20 mV, respectively. Drug encapsulation efficiency was 90%. NMFTC was readily taken up by human monocyte-derived macrophages yielding readily detected intracellular FTC triphosphates and an extended PK profile. CONCLUSION: NMFTC shows improved antiretroviral activities over native FTC. This is coordinate with its extended apparent half-life. The work represents an incremental advance in the development of a long-acting FTC formulation.


Asunto(s)
Composición de Medicamentos , Emtricitabina/farmacología , Nanopartículas/química , Profármacos/farmacología , Animales , Antirretrovirales/farmacología , Espectroscopía de Resonancia Magnética con Carbono-13 , Emtricitabina/sangre , Emtricitabina/síntesis química , Emtricitabina/química , Humanos , Cinética , Macrófagos/efectos de los fármacos , Masculino , Nanopartículas/ultraestructura , Profármacos/síntesis química , Profármacos/química , Espectroscopía de Protones por Resonancia Magnética , Ratas Sprague-Dawley
14.
J Control Release ; 311-312: 201-211, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31491432

RESUMEN

Antiretroviral therapy requires lifelong daily dosing to attain viral suppression, restore immune function, and improve quality of life. As a treatment alternative, long-acting (LA) antiretrovirals can sustain therapeutic drug concentrations in blood for prolonged time periods. The success of recent clinical trials for LA parenteral cabotegravir and rilpivirine highlight the emergence of these new therapeutic options. Further optimization can improve dosing frequency, lower injection volumes, and facilitate drug-tissue distributions. To this end, we report the synthesis of a library of RPV prodrugs designed to sustain drug plasma concentrations and improved tissue biodistribution. The lead prodrug M3RPV was nanoformulated into the stable LA injectable NM3RPV. NM3RPV treatment led to RPV plasma concentrations above the protein-adjusted 90% inhibitory concentration for 25 weeks with substantial tissue depots after a single intramuscular injection in BALB/cJ mice. NM3RPV elicited 13- and 26-fold increases in the RPV apparent half-life and mean residence time compared to native drug formulation. Taken together, proof-of-concept is provided that nanoformulated RPV prodrugs can extend the apparent drug half-life and improve tissue biodistribution. These results warrant further development for human use.


Asunto(s)
Fármacos Anti-VIH/administración & dosificación , Nanopartículas/administración & dosificación , Profármacos/administración & dosificación , Rilpivirina/administración & dosificación , Animales , Fármacos Anti-VIH/farmacocinética , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/farmacocinética , VIH-1/efectos de los fármacos , Humanos , Macaca mulatta , Macrófagos/metabolismo , Masculino , Ratones Endogámicos BALB C , Profármacos/farmacocinética , Rilpivirina/farmacocinética , Distribución Tisular
15.
Biomaterials ; 185: 174-193, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30245386

RESUMEN

Antiretroviral therapy (ART) has changed the outcome of human immunodeficiency virus type one (HIV-1) infection from certain death to a life free of disease co-morbidities. However, infected people must remain on life-long daily ART. ART reduces but fails to eliminate the viral reservoir. In order to improve upon current treatment regimens, our laboratory created long acting slow effective release (LASER) ART nanoformulated prodrugs from native medicines. LASER ART enables antiretroviral drugs (ARVs) to better reach target sites of HIV-1 infection while, at the same time, improve ART's half-life and potency. However, novel ARV design has been slowed by prolonged pharmacokinetic testing requirements. To such ends, tri-modal theranostic nanoparticles were created with single-photon emission computed tomography (SPECT/CT), magnetic resonance imaging (MRI) and fluorescence capabilities to predict LASER ART biodistribution. The created theranostic ARV probes were then employed to monitor drug tissue distribution and potency. Intrinsically 111Indium (111In) radiolabeled, europium doped cobalt-ferrite particles and rilpivirine were encased in a polycaprolactone core surrounded by a lipid shell (111InEuCF-RPV). Particle cell and tissue distribution, and antiretroviral activities were sustained in macrophage tissue depots. 111InEuCF-PCL/RPV particles injected into mice demonstrated co-registration of MRI and SPECT/CT tissue signals with RPV and cobalt. Cell and animal particle biodistribution paralleled ARV activities. We posit that particle selection can predict RPV distribution and potency facilitated by multifunctional theranostic nanoparticles.


Asunto(s)
Antirretrovirales/farmacocinética , Nanopartículas/química , Rilpivirina/farmacocinética , Animales , Antirretrovirales/farmacología , Cobalto/química , Sistemas de Liberación de Medicamentos , Europio/química , Compuestos Férricos/química , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Imagen por Resonancia Magnética/métodos , Masculino , Ratones Endogámicos BALB C , Imagen Óptica/métodos , Rilpivirina/farmacología , Nanomedicina Teranóstica , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único/métodos
16.
J Biomed Mater Res A ; 106(11): 2881-2890, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30369055

RESUMEN

An urgent need to deliver therapeutics across the blood-brain barrier (BBB) underlies a paucity of effective therapies currently available for treatment of degenerative, infectious, traumatic, chemical, and metabolic disorders of the nervous system. With an eye toward achieving this goal, an in vitro BBB model was employed to simulate biodegradable polyanhydride nanoparticle-based drug delivery to the brain. Using a combination of confocal microscopy, flow cytometry, and high performance liquid chromatography, we examined the potential of polyanhydride nanoparticles containing the anti-oxidant, mito-apocynin, to be internalized and then transferred from monocytes to human brain microvascular endothelial cells. The efficacy of this nanoparticle-based delivery platform was demonstrated by neuronal protection against oxidative stress. Taken together, this polyanhydride nanoparticle-based delivery system holds promise for enhancing neuroprotection by facilitating drug transport across the BBB. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2881-2890, 2018.


Asunto(s)
Antioxidantes/administración & dosificación , Barrera Hematoencefálica/metabolismo , Portadores de Fármacos/metabolismo , Nanopartículas/metabolismo , Polianhídridos/metabolismo , Adulto , Antioxidantes/farmacocinética , Transporte Biológico , Encéfalo/metabolismo , Células Cultivadas , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Células Endoteliales/metabolismo , Humanos , Monocitos/metabolismo , Nanopartículas/química , Nanopartículas/ultraestructura , Polianhídridos/química , Puntos Cuánticos/química , Puntos Cuánticos/metabolismo
17.
Theranostics ; 8(1): 256-276, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29290806

RESUMEN

RATIONALE: Long-acting slow effective release antiretroviral therapy (LASER ART) was developed to improve patient regimen adherence, prevent new infections, and facilitate drug delivery to human immunodeficiency virus cell and tissue reservoirs. In an effort to facilitate LASER ART development, "multimodal imaging theranostic nanoprobes" were created. These allow combined bioimaging, drug pharmacokinetics and tissue biodistribution tests in animal models. METHODS: Europium (Eu3+)- doped cobalt ferrite (CF) dolutegravir (DTG)- loaded (EuCF-DTG) nanoparticles were synthesized then fully characterized based on their size, shape and stability. These were then used as platforms for nanoformulated drug biodistribution. RESULTS: Folic acid (FA) decoration of EuCF-DTG (FA-EuCF-DTG) nanoparticles facilitated macrophage targeting and sped drug entry across cell barriers. Macrophage uptake was higher for FA-EuCF-DTG than EuCF-DTG nanoparticles with relaxivities of r2 = 546 mM-1s-1 and r2 = 564 mM-1s-1 in saline, and r2 = 850 mM-1s-1 and r2 = 876 mM-1s-1 in cells, respectively. The values were ten or more times higher than what was observed for ultrasmall superparamagnetic iron oxide particles (r2 = 31.15 mM-1s-1 in saline) using identical iron concentrations. Drug particles were detected in macrophage Rab compartments by dual fluorescence labeling. Replicate particles elicited sustained antiretroviral responses. After parenteral injection of FA-EuCF-DTG and EuCF-DTG into rats and rhesus macaques, drug, iron and cobalt levels, measured by LC-MS/MS, magnetic resonance imaging, and ICP-MS were coordinate. CONCLUSION: We posit that these theranostic nanoprobes can assess LASER ART drug delivery and be used as part of a precision nanomedicine therapeutic strategy.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Imagen Multimodal/métodos , Nanomedicina Teranóstica/métodos , Animales , Sistemas de Liberación de Medicamentos/métodos , Europio/química , Europio/farmacocinética , Ácido Fólico/química , Compuestos Heterocíclicos con 3 Anillos/química , Compuestos Heterocíclicos con 3 Anillos/farmacocinética , Macaca mulatta , Macrófagos/metabolismo , Microscopía Confocal , Nanopartículas/química , Oxazinas , Piperazinas , Piridonas
18.
Toxicology ; 230(2-3): 234-43, 2007 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-17161896

RESUMEN

The industrial solvent trichloroethylene (TCE) and its major metabolites have been shown to cause formic aciduria in male rats. We have examined whether chloral hydrate (CH) and trichloroacetic acid (TCA), known metabolites of TCE, produce an increase in formic acid in vitro in cultures of rat hepatocytes or human renal proximal tubule cells (HRPTC). The metabolism and cytotoxicity of CH was also examined to establish that the cells were metabolically active and not compromised by toxicity. Rat hepatocytes and HRPTC were cultured in serum-free medium and then treated with 0.3-3mM CH for 3 days or 0.03-3mM CH for 10 days, respectively and formic acid production, metabolism to trichloroethanol (TCE-OH) and TCA and cytotoxicity determined. No increase in formic acid production in rat hepatocytes or HRPTC exposed to CH was observed over and above that due to chemical degradation, neither was formic acid production observed in rat hepatocytes exposed to TCA. HRPTC metabolized CH to TCE-OH and TCA with a 12-fold greater capacity to form TCE-OH versus TCA. Rat hepatocytes exhibited a 1.6-fold and three-fold greater capacity than HRPTC to form TCE-OH and TCA, respectively. CH and TCA were not cytotoxic to rat hepatocytes at concentrations up to 3mM/day for 3 days. With HRPTC, one sample showed no cytotoxicity to CH at concentrations up to 3mM/day for 10 days, while in another cytotoxicity was seen at 1mM/day for 3 days. In summary, increased formic acid production was not observed in rat hepatocytes or HRPTC exposed to TCE metabolites, suggesting that the in vivo response cannot be modelled in vitro. CH was toxic to HRPTC at millimolar concentrations/day over 10 days, while glutathione derived metabolites of TCE were toxic at micromolar concentrations/day over 10 days [Lock, E.A., Reed, C.J., 2006. Trichloroethylene: mechanisms of renal toxicity and renal cancer and relevance to risk assessment. Toxicol. Sci. 19, 313-331] supporting the view that glutathione derived metabolites are likely to be responsible for nephrotoxicity.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Hidrato de Cloral/toxicidad , Formiatos/metabolismo , Hepatocitos/efectos de los fármacos , Enfermedades Renales/inducido químicamente , Túbulos Renales Proximales/efectos de los fármacos , Ácido Tricloroacético/toxicidad , Adolescente , Adulto , Animales , Cromatografía de Gases , Etilenclorhidrina/análogos & derivados , Etilenclorhidrina/metabolismo , Hepatocitos/metabolismo , Humanos , Enfermedades Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Hepatopatías/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Ratas
19.
Acta Biomater ; 49: 507-520, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27916740

RESUMEN

The size, shape and chemical composition of europium (Eu3+) cobalt ferrite (CFEu) nanoparticles were optimized for use as a "multimodal imaging nanoprobe" for combined fluorescence and magnetic resonance bioimaging. Doping Eu3+ ions into a CF structure imparts unique bioimaging and magnetic properties to the nanostructure that can be used for real-time screening of targeted nanoformulations for tissue biodistribution assessment. The CFEu nanoparticles (size ∼7.2nm) were prepared by solvothermal techniques and encapsulated into poloxamer 407-coated mesoporous silica (Si-P407) to form superparamagnetic monodisperse Si-CFEu nanoparticles with a size of ∼140nm. Folic acid (FA) nanoparticle decoration (FA-Si-CFEu, size ∼140nm) facilitated monocyte-derived macrophage (MDM) targeting. FA-Si-CFEu MDM uptake and retention was higher than seen with Si-CFEu nanoparticles. The transverse relaxivity of both Si-CFEu and FA-Si-CFEu particles were r2=433.42mM-1s-1 and r2=419.52mM-1s-1 (in saline) and r2=736.57mM-1s-1 and r2=814.41mM-1s-1 (in MDM), respectively. The results were greater than a log order-of-magnitude than what was observed at replicate iron concentrations for ultrasmall superparamagnetic iron oxide (USPIO) particles (r2=31.15mM-1s-1 in saline) and paralleled data sets obtained for T2 magnetic resonance imaging. We now provide a developmental opportunity to employ these novel particles for theranostic drug distribution and efficacy evaluations. STATEMENT OF SIGNIFICANCE: A novel europium (Eu3+) doped cobalt ferrite (Si-CFEu) nanoparticle was produced for use as a bioimaging probe. Its notable multifunctional, fluorescence and imaging properties, allows rapid screening of future drug biodistribution. Decoration of the Si-CFEu particles with folic acid increased its sensitivity and specificity for magnetic resonance imaging over a more conventional ultrasmall superparamagnetic iron oxide particles. The future use of these particles in theranostic tests will serve as a platform for designing improved drug delivery strategies to combat inflammatory and infectious diseases.


Asunto(s)
Cobalto/química , Europio/química , Compuestos Férricos/química , Imagen por Resonancia Magnética , Nanopartículas/química , Dióxido de Silicio/química , Animales , Endocitosis , Ácido Fólico/química , Humanos , Inmunohistoquímica , Macrófagos/metabolismo , Macrófagos/ultraestructura , Masculino , Microscopía de Fuerza Atómica , Microscopía Confocal , Monocitos/citología , Nanopartículas/toxicidad , Ratas Sprague-Dawley , Espectroscopía Infrarroja por Transformada de Fourier , Termogravimetría , Distribución Tisular
20.
Environ Health Perspect ; 114(8): 1237-42, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16882532

RESUMEN

BACKGROUND: Trichloroethylene (TCE) is a suspected human carcinogen and a common groundwater contaminant. Chloral hydrate (CH) is the major metabolite of TCE formed in the liver by cytochrome P450 2E1. CH is metabolized to the hepatocarcinogen trichloroacetate (TCA) by aldehyde dehydrogenase (ALDH) and to the noncarcinogenic metabolite trichloroethanol (TCOH) by alcohol dehydrogenase (ADH). ALDH and ADH are polymorphic in humans, and these polymorphisms are known to affect the elimination of ethanol. It is therefore possible that polymorphisms in CH metabolism will yield subpopulations with greater than expected TCA formation with associated enhanced risk of liver tumors after TCE exposure. METHODS: The present studies were undertaken to determine the feasibility of using commercially available, cryogenically preserved human hepatocytes to determine simultaneously the kinetics of CH metabolism and ALDH/ADH genotype. Thirteen human hepatocyte samples were examined. Linear reciprocal plots were obtained for 11 ADH and 12 ALDH determinations. RESULTS: There was large interindividual variation in the Vmax values for both TCOH and TCA formation. Within this limited sample size, no correlation with ADH/ALDH genotype was apparent. Despite the large variation in Vmax values among individuals, disposition of CH into the two competing pathways was relatively constant. CONCLUSIONS: These data support the use of cryopreserved human hepatocytes as an experimental system to generate metabolic and genomic information for incorporation into TCE cancer risk assessment models. The data are discussed with regard to cellular factors, other than genotype, that may contribute to the observed variability in metabolism of CH in human liver.


Asunto(s)
Hidrato de Cloral/metabolismo , Etilenclorhidrina/análogos & derivados , Hepatocitos/efectos de los fármacos , Ácido Tricloroacético/metabolismo , Ácido Tricloroacético/toxicidad , Tricloroetileno/metabolismo , Tricloroetileno/toxicidad , Adulto , Anciano , Alcohol Deshidrogenasa/genética , Alcohol Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Animales , Criopreservación , Etilenclorhidrina/metabolismo , Etilenclorhidrina/toxicidad , Femenino , Genotipo , Humanos , Técnicas In Vitro , Cinética , Masculino , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Medición de Riesgo , Solventes/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA