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













Base de datos
Intervalo de año de publicación
1.
Drug Metab Dispos ; 50(6): 781-797, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34154993

RESUMEN

Conjugation of oligonucleotide therapeutics, including small interfering RNAs (siRNAs) or antisense oligonucleotides, to N-acetylgalactosamine (GalNAc) ligands has become the primary strategy for hepatocyte-targeted delivery, and with the recent approvals of GIVLAARI (givosiran) for the treatment of acute hepatic porphyria, OXLUMO (lumasiran) for the treatment of primary hyperoxaluria, and Leqvio (inclisiran) for the treatment of hypercholesterolemia, the technology has been well validated clinically. Although much knowledge has been gained over decades of development, there is a paucity of published literature on the drug metabolism and pharmacokinetic properties of GalNAc-siRNA. With this in mind, the goals of this minireview are to provide an aggregate analysis of these nonclinical absorption, distribution, metabolism, and excretion (ADME) data to build confidence on the translation of these properties to human. Upon subcutaneous administration, GalNAc-conjugated siRNAs are quickly distributed to the liver, resulting in plasma pharmacokinetic (PK) properties that reflect rapid elimination through asialoglycoprotein receptor-mediated uptake from circulation into hepatocytes. These studies confirm that liver PK, including half-life and, most importantly, siRNA levels in RNA-induced silencing complex in hepatocytes, are better predictors of pharmacodynamics (PD) than plasma PK. Several in vitro and in vivo nonclinical studies were conducted to characterize the ADME properties of GalNAc-conjugated siRNAs. These studies demonstrate that the PK/PD and ADME properties of GalNAc-conjugated siRNAs are highly conserved across species, are largely predictable, and can be accurately scaled to human, allowing us to identify efficacious and safe clinical dosing regimens in the absence of human liver PK profiles. SIGNIFICANCE STATEMENT: Several nonclinical ADME studies have been conducted in order to provide a comprehensive overview of the disposition and elimination of GalNAc-conjugated siRNAs and the pharmacokinetic/pharmacodynamic translation between species. These studies demonstrate that the ADME properties of GalNAc-conjugated siRNAs are well correlated and predictable across species, building confidence in the ability to extrapolate to human.


Asunto(s)
Acetilgalactosamina , Porfirias Hepáticas , Acetilgalactosamina/farmacocinética , Receptor de Asialoglicoproteína/metabolismo , Hepatocitos/metabolismo , Humanos , Porfirias Hepáticas/metabolismo , ARN Interferente Pequeño/genética
2.
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
3.
Drug Metab Dispos ; 49(7): 572-580, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33941543

RESUMEN

Givosiran is an N-acetylgalactosamine-conjugated RNA interference therapeutic that targets 5'-aminolevulinate synthase 1 mRNA in the liver and is currently marketed for the treatment of acute hepatic porphyria. Herein, nonclinical pharmacokinetics and absorption, distribution, metabolism, and excretion properties of givosiran were characterized. Givosiran was completely absorbed after subcutaneous administration with relatively short plasma elimination half-life (t1/2; less than 4 hours). Plasma exposure increased approximately dose proportionally with no accumulation after repeat doses. Plasma protein binding was concentration dependent across all species tested and was around 90% at clinically relevant concentration in human. Givosiran predominantly distributed to the liver by asialoglycoprotein receptor-mediated uptake, and the t1/2 in the liver was significantly longer (∼1 week). Givosiran was metabolized by nucleases, not cytochrome P450 (P450) isozymes, across species with no human unique metabolites. Givosiran metabolized to form one primary active metabolite with the loss of one nucleotide from the 3' end of antisense strand, AS(N-1)3' givosiran, which was equipotent to givosiran. Renal and fecal excretion were minor routes of elimination of givosiran as approximately 10% and 16% of the dose was recovered intact in excreta of rats and monkeys, respectively. Givosiran is not a substrate, inhibitor, or inducer of P450 isozymes, and it is not a substrate or inhibitor of uptake and most efflux transporters. Thus, givosiran has a low potential of mediating drug-drug interactions involving P450 isozymes and drug transporters. SIGNIFICANCE STATEMENT: Nonclinical pharmacokinetics and absorption, distribution, metabolism, and excretion (ADME) properties of givosiran were characterized. Givosiran shows similar pharmacokinetics and ADME properties across rats and monkeys in vivo and across human and animal matrices in vitro. Subcutaneous administration results in adequate exposure of givosiran to the target organ (liver). These studies support the interpretation of toxicology studies, help characterize the disposition of givosiran in humans, and support the clinical use of givosiran for the treatment of acute hepatic porphyria.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Pirrolidinas/farmacocinética , 5-Aminolevulinato Sintetasa/antagonistas & inhibidores , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/farmacocinética , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Femenino , Semivida , Inyecciones Subcutáneas , Eliminación Intestinal , Macaca fascicularis , Masculino , Modelos Animales , Porfobilinógeno Sintasa/deficiencia , Porfirias Hepáticas/tratamiento farmacológico , Pirrolidinas/administración & dosificación , Ratas , Eliminación Renal , Distribución Tisular
4.
Drugs ; 81(7): 841-848, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33871817

RESUMEN

Givosiran (Givlaari®) is an δ-aminolevulinic acid synthase 1 (ALAS1)-directed small interfering RNA (siRNA) approved for the treatment of acute hepatic porphyria (AHP). In the phase 3 ENVISION trial, givosiran significantly reduced the annualized rate of composite porphyria attacks (i.e. attacks requiring hospitalization, urgent healthcare visit or intravenous hemin administration at home) compared with placebo in patients with recurrent acute intermittent porphyria (the most common type of AHP) attacks. Givosiran also improved several other outcomes, including hemin use and pain (the cardinal symptom of AHP). While generally well tolerated with an acceptable safety profile, the drug may increase the risk of hepatic and kidney adverse events. Givosiran offers the convenience of once-monthly subcutaneous administration. Available evidence indicates that givosiran is an important newer therapeutic option for patients with AHP and severe recurrent attacks.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Porfobilinógeno Sintasa/deficiencia , Porfiria Intermitente Aguda/tratamiento farmacológico , Porfirias Hepáticas/tratamiento farmacológico , Pirrolidinas/farmacología , Pirrolidinas/uso terapéutico , 5-Aminolevulinato Sintetasa/antagonistas & inhibidores , Acetilgalactosamina/efectos adversos , Acetilgalactosamina/farmacocinética , Acetilgalactosamina/farmacología , Acetilgalactosamina/uso terapéutico , Lesión Renal Aguda/inducido químicamente , Enfermedad Hepática Inducida por Sustancias y Drogas , Interacciones Farmacológicas , Hemina/administración & dosificación , Hospitalización , Humanos , Dolor/tratamiento farmacológico , Dolor/etiología , Porfiria Intermitente Aguda/complicaciones , Porfirias Hepáticas/complicaciones , Pirrolidinas/efectos adversos , Pirrolidinas/farmacocinética , ARN Interferente Pequeño , Ensayos Clínicos Controlados Aleatorios como Asunto , Índice de Severidad de la Enfermedad
5.
Clin Pharmacol Ther ; 108(1): 63-72, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31994716

RESUMEN

Givosiran is a small interfering ribonucleic acid agent that was recently approved in the United States for the treatment of acute hepatic porphyria (AHP). This phase I study evaluated the safety, pharmacokinetic, and pharmacodynamic profile of subcutaneously (SC) administered givosiran in patients with acute intermittent porphyria, the most common AHP type. Givosiran was rapidly absorbed from the SC injection site with peak plasma concentrations achieved within 0.5-5 hours followed by elimination with a short half-life of 4-10 hours. Plasma exposures of AS(N-1)3' givosiran, an active metabolite with equal potency as givosiran, was 35%-75%. Givosiran treatment resulted in a rapid and dose-dependent reduction in urinary aminolevulinic acid (ALA) and porphobilinogen (PBG) towards the upper limit of normal (ULN) in AHP patients. Greater and more sustained reductions in ALA and PBG were achieved with once monthly dosing compared with once quarterly dosing. After monthly dosing, trough ALA levels were reduced to below the ULN, approximately 95% reduction from baseline, at both the 2.5 and 5.0 mg/kg doses.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Ácido Aminolevulínico/orina , Porfobilinógeno/orina , Porfiria Intermitente Aguda/tratamiento farmacológico , Pirrolidinas/administración & dosificación , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/farmacocinética , Acetilgalactosamina/farmacología , Adulto , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Semivida , Humanos , Inyecciones Subcutáneas , Masculino , Persona de Mediana Edad , Pirrolidinas/farmacocinética , Pirrolidinas/farmacología , Adulto Joven
6.
Mol Diagn Ther ; 24(1): 61-68, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31792921

RESUMEN

In November 2019 givosiran became the second small interfering RNA (siRNA)-based drug to receive US Food and Drug Administration (FDA) approval, it has been developed for the treatment of acute intermittent porphyria (AIP), a disorder characterized by life-threatening acute neurovisceral attacks. The porphyrias are a group of disorders in which enzymatic deficiencies in heme production lead to toxic accumulation of delta-aminolevulinic acid (ALA) and porphobilinogen (PBG), which are involved in the neurovisceral attacks. Givosiran acts as a conventional siRNA to trigger RNA interference (RNAi)-mediated gene silencing on delta-ALA synthase 1 (ALAS1), thus returning ALA and PBG metabolites to the physiological level to attenuate further neurotoxicity. Givosiran makes use of a new hepatic-delivery system that conjugates three GalNac (N-acetylgalactosamine) molecules to the siRNA passenger strand. GalNac binds to the liver asialoglycoprotein receptor, favoring the internalization of these GalNac-conjugated siRNAs into the hepatic cells. In a phase I study, subcutaneous monthly administration of givosiran 2.5 mg/kg reduced > 90% of ALA and PBG content. This siRNA is being analyzed in ENVISION (NCT03338816), a phase III, multicenter, placebo-controlled randomized controlled trial. In preliminary results, givosiran achieved clinical endpoints for AIP, reducing urinary ALA levels, and presented a safety profile that enabled further drug development. The clinical performance of givosiran revealed that suppression of ALAS1 by GalNac-decorated siRNAs represents an additional approach for the treatment of patients with AIP that manifests recurrent acute neurovisceral attacks.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Silenciador del Gen , Terapia Genética , Óxido Nítrico Sintasa de Tipo I/genética , Pirrolidinas/uso terapéutico , Interferencia de ARN , ARN Interferente Pequeño/uso terapéutico , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/efectos adversos , Acetilgalactosamina/farmacocinética , Acetilgalactosamina/uso terapéutico , Ensayos Clínicos como Asunto , Desarrollo de Medicamentos , Terapia Genética/efectos adversos , Terapia Genética/métodos , Hemo/biosíntesis , Humanos , Porfiria Intermitente Aguda , Pirrolidinas/administración & dosificación , Pirrolidinas/efectos adversos , Pirrolidinas/farmacocinética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/efectos adversos , ARN Interferente Pequeño/farmacocinética , Resultado del Tratamiento
7.
Drug Metab Dispos ; 47(10): 1174-1182, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31097425

RESUMEN

Understanding small interfering RNA (siRNA) fraction unbound (f u) in relevant physiologic compartments is critical for establishing pharmacokinetic-pharmacodynamic relationships for this emerging modality. In our attempts to isolate the equilibrium free fraction of N-acetylgalactosamine-conjugated siRNA using classic small-molecule in vitro techniques, we found that the hydrodynamic radius was critical in determining the size exclusion limit requirements for f u isolation, largely validating the siRNA "rigid rod" hypothesis. With this knowledge, we developed an orthogonally validated 50 kDa molecular-mass cutoff ultrafiltration assay to quantify f u in biologic matrices including human, nonhuman primate, rat, and mouse plasma, and human liver homogenate. To enhance understanding of the siRNA-plasma interaction landscape, we examined the effects of various common oligonucleotide therapeutic modifications to the ribose and helix backbone on siRNA f u in plasma (f u,plasma) and found that chemical modifications can alter plasma protein binding by at least 20%. Finally, to gain insight into which specific plasma proteins bind to siRNA, we developed a qualitative screen to identify binding "hits" across a panel of select purified human plasma proteins.


Asunto(s)
Acetilgalactosamina/farmacocinética , Proteínas Sanguíneas/metabolismo , Hígado/metabolismo , ARN Interferente Pequeño/farmacocinética , Acetilgalactosamina/química , Adulto , Animales , Femenino , Humanos , Macaca fascicularis , Ratones , Unión Proteica , ARN Interferente Pequeño/química , Ratas
8.
Nucleic Acid Ther ; 29(1): 16-32, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30570431

RESUMEN

Advances in medicinal chemistry have produced new chemical classes of antisense oligonucleotides (ASOs) with enhanced therapeutic properties. Conjugation of the triantennary N-acetylgalactosamine (GalNAc3) moiety to the extensively characterized phosphorothioate (PS)-modified 2'-O-methoxyethyl (2'MOE) ASO exemplifies such an advance. This structure-activity optimized moiety effects receptor-mediated uptake of the ASO prodrug through the asialoglycoprotein receptor 1 to support selective targeting of RNAs expressed by hepatocytes. In this study we report the integrated assessment of data available from randomized placebo-controlled dose-ranging studies of this chemical class of ASOs administered systemically to healthy human volunteers. First, we compare the pharmacokinetic and pharmacodynamic profiles of a subset of the GalNAc3-conjugated PS-modified 2'MOE ASOs to the parent PS-modified 2'MOE ASOs for which plasma analytes are available. We then evaluate the safety profile of the full set of GalNAc3-conjugated PS-modified 2'MOE ASO conjugates by the incidence of signals in standardized laboratory tests and by the mean laboratory test results as a function of dose level over time. With hepatocyte targeted delivery, the ED50 for the GalNAc3-conjugated PS-modified 2'MOE ASO subset ranges from 4 to 10 mg/week, up to 30-fold more potent than the parent PS-modified 2'MOE ASO. No GalNAc3-conjugated PS-modified 2'MOE ASO class effects were identified from the assessment of the integrated laboratory test data across all doses tested with either single or multidose regimens. The increase in potency supports an increase in the safety margin for this new chemical class of ASOs now under broad investigation in the clinic. Although the total exposure is limited in the initial phase 1 trials, ongoing and future investigations in patient populations will support evaluation of the effects of long-term exposure.


Asunto(s)
Acetilgalactosamina/administración & dosificación , Receptor de Asialoglicoproteína/genética , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Fosforotioatos/administración & dosificación , Acetilgalactosamina/sangre , Acetilgalactosamina/farmacocinética , Receptor de Asialoglicoproteína/sangre , Biomarcadores Farmacológicos/sangre , Relación Dosis-Respuesta a Droga , Femenino , Voluntarios Sanos , Hepatocitos/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Oligonucleótidos Antisentido/sangre , Oligonucleótidos Antisentido/farmacocinética , Oligonucleótidos Fosforotioatos/sangre , Oligonucleótidos Fosforotioatos/farmacocinética , ARN/antagonistas & inhibidores , ARN/sangre , ARN/genética , Relación Estructura-Actividad
9.
J Control Release ; 266: 310-320, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-28987883

RESUMEN

RNA interference (RNAi) has the potential to reversibly silence any gene with high efficiency and specificity. To fulfill the clinical potential of RNAi, delivery vehicles are required to transport the short interfering RNA (siRNA) to the site of action in the cells of target tissues. Here, we describe the features of novel liver-targeted siRNA nanoparticles (NPs), co-assembled due to the complexation of alginate sulfate (AlgS) with siRNA, mediated by calcium ions bridges (AlgS-Ca2+-siRNA NPs) and then bioconjugation of a targeting ligand onto the AlgS upon the NP surface. To gain insight into the complexation process and confirm AlgS accessibility on NP surface, we investigated different schemes for fabrication. All resulting NPs, independently of the component addition order, were of average size of 130-150nm, had surface charge of <-10mV, exhibited a similar atomic composition on their surface, were efficiently uptaken by HepG2 cells and induced approx. ~90% silencing of STAT3 gene. Ca2+ and AlgS concentrations in NPs affected cell uptake and gene silencing. Bioconjugation of N-acetylgalactosamine (GalNAc), a ligand to the asialoglycoprotein receptor (ASGPR) overexpressed on hepatocytes, was validated by XPS analysis and cell uptake by receptor-mediated mechanism. After intravenous (i.v.) injection to BALB/c mice, GalNAc-NPs were targeted to liver by a factor of ~3 with lesser renal clearance compared to non-targeted NPs. We foresee that the combined advantages of site-specific targeting and reversibility of the tri-component NPs as well as the simplicity of their fabrication make them an attractive system for targeted delivery of siRNA.


Asunto(s)
Acetilgalactosamina/administración & dosificación , Hígado/metabolismo , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Acetilgalactosamina/química , Acetilgalactosamina/farmacocinética , Alginatos/administración & dosificación , Alginatos/química , Alginatos/farmacocinética , Animales , Calcio/administración & dosificación , Calcio/química , Calcio/farmacocinética , Supervivencia Celular , Femenino , Silenciador del Gen , Células Hep G2 , Humanos , Ratones Endogámicos BALB C , Nanopartículas/química , ARN Interferente Pequeño/farmacocinética , Electricidad Estática
10.
Nucleic Acids Res ; 45(19): 10969-10977, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28981809

RESUMEN

Covalent attachment of a synthetic triantennary N-acetylagalactosamine (GalNAc) ligand to chemically modified siRNA has enabled asialoglycoprotein (ASGPR)-mediated targeted delivery of therapeutically active siRNAs to hepatocytes in vivo. This approach has become transformative for the delivery of RNAi therapeutics as well as other classes of investigational oligonucleotide therapeutics to the liver. For efficient functional delivery of intact drug into the desired subcellular compartment, however, it is critical that the nucleic acids are stabilized against nucleolytic degradation. Here, we compared two siRNAs of the same sequence but with different modification pattern resulting in different degrees of protection against nuclease activity. In vitro stability studies in different biological matrices show that 5'-exonuclease is the most prevalent nuclease activity in endo-lysosomal compartments and that additional stabilization in the 5'-regions of both siRNA strands significantly enhances the overall metabolic stability of GalNAc-siRNA conjugates. In good agreement with in vitro findings, the enhanced stability translated into substantially improved liver exposure, gene silencing efficacy and duration of effect in mice. Follow-up studies with a second set of conjugates targeting a different transcript confirmed the previous results, provided additional insights into kinetics of RISC loading and demonstrated excellent translation to non-human primates.


Asunto(s)
Acetilgalactosamina/farmacocinética , Riñón/metabolismo , Hígado/metabolismo , ARN Interferente Pequeño/farmacocinética , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/metabolismo , Animales , Área Bajo la Curva , Sistemas de Liberación de Medicamentos/métodos , Humanos , Hígado/citología , Masculino , Tasa de Depuración Metabólica , Ratones Endogámicos C57BL , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/metabolismo
11.
Nucleic Acid Ther ; 26(6): 372-380, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27500733

RESUMEN

Triantennary N-acetyl galactosamine (GalNAc3)-conjugated antisense oligonucleotides (ASOs) have greatly improved potency due to receptor-mediated uptake into hepatocyte. The disposition and pharmacokinetics of ISIS 681257, a GalNAc3-conjugated ASO, were studied in monkeys. Following subcutaneous (SC) injection, ISIS 681257 was rapidly absorbed into the systemic circulation, with peak plasma levels observed within hours after dosing. After reaching Cmax, plasma concentrations rapidly declined in a multiexponential manner and were characterized by a dominant initial rapid distribution phase in which drug transferred to tissues from circulation, followed by a much slower terminal elimination phase (half-life of 4 weeks). Intact ISIS 681257 is the major full-length oligonucleotide species in plasma (≥70%). In tissues, the conjugated-GalNAc sugar moiety was rapidly metabolized, leaving the fully unconjugated form as the only full-length oligonucleotide detected at 48 h after dosing. Unconjugated ISIS 681257 cleared slowly from tissues with a half-life of 4 weeks. ISIS 681257 was highly bound to plasma proteins (>97% bound), which limited its urinary excretion. Disposition of ISIS 681257 in plasma and liver appeared nonlinear over the 1-40 mg/kg dose range studied. The plasma and liver tissue concentration data were well described by a population based mixed-effects modeling approach with Michaelis-Menten uptake from plasma to liver. Safety data from the study and the good exposure, as well as the extended half-life of the unconjugated ASO in the liver, support further development and less frequent dosing in Phase I clinical study.


Asunto(s)
Acetilgalactosamina/farmacocinética , Glicoconjugados/farmacocinética , Lipoproteína(a)/metabolismo , Oligonucleótidos Antisentido/farmacocinética , Oligonucleótidos Fosforotioatos/farmacocinética , Acetilgalactosamina/metabolismo , Animales , Biotransformación , Proteínas Sanguíneas/metabolismo , Femenino , Glicoconjugados/metabolismo , Semivida , Hepatocitos/metabolismo , Inyecciones Subcutáneas , Hígado/metabolismo , Masculino , Oligonucleótidos Antisentido/metabolismo , Oligonucleótidos Fosforotioatos/metabolismo , Unión Proteica , División del ARN
12.
J Agric Food Chem ; 64(12): 2604-11, 2016 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-26975338

RESUMEN

The ß-galactosidases from Lactobacillus reuteri L103 (Lreußgal), Lactobacillus delbrueckii subsp. bulgaricus DSM 20081 (Lbulßgal), and Bifidobacterium breve DSM 20281 (Bbreßgal-I and Bbreßgal-II) were investigated in detail with respect to their propensity to transfer galactosyl moieties onto lactose, its hydrolysis products D-glucose and D-galactose, and certain sugar acceptors such as N-acetyl-D-glucosamine (GlcNAc), N-acetyl-D-galactosamine (GalNAc), and L-fucose (Fuc) under defined, initial velocity conditions. The rate constants or partitioning ratios (kNu/kwater) determined for these different acceptors (termed nucleophiles, Nu) were used as a measure for the ability of a certain substance to act as a galactosyl acceptor of these ß-galactosidases. When using Lbulßgal or Bbreßgal-II, the galactosyl transfer to GlcNAc was 6 and 10 times higher than that to lactose, respectively. With lactose and GlcNAc used in equimolar substrate concentrations, Lbulßgal and Bbreßgal-II catalyzed the formation of N-acetyl-allolactosamine with the highest yields of 41 and 24%, respectively, as calculated from the initial GlcNAc concentration.


Asunto(s)
Galactosa/metabolismo , Lactobacillus/enzimología , Lactosa/metabolismo , Oligosacáridos/metabolismo , Transferasas/metabolismo , beta-Galactosidasa/metabolismo , Acetilgalactosamina/análogos & derivados , Acetilgalactosamina/metabolismo , Acetilgalactosamina/farmacocinética , Acetilglucosamina/metabolismo , Acetilglucosamina/farmacocinética , Bifidobacterium/metabolismo , Galactosamina/metabolismo , Glucosamina/metabolismo , Glucosa/metabolismo , Limosilactobacillus reuteri/metabolismo , Especificidad por Sustrato
13.
Bioconjug Chem ; 24(12): 2088-103, 2013 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-24147780

RESUMEN

Dendrimer clearing agents represent a unique class of compounds for use in multistep targeting (MST) in radioimmunotherapy and imaging. These compounds were developed to facilitate the removal of excess tumor-targeting monoclonal antibody (mAb) prior to administration of the radionuclide to minimize exposure of normal tissue to radiation. Clearing agents are designed to capture the circulating mAb, and target it to the liver for metabolism. Glycodendrons are ideally suited for MST applications as these highly branched compounds are chemically well-defined, thus advantageous over heterogeneous macromolecules. Previous studies have described glycodendron 3 as a clearing agent for use in three-step MST protocols, and early in vivo assessment of 3 showed promise. However, synthetic challenges have hampered its availability for further development. In this report we describe a new sequence of chemical steps which enables the straightforward synthesis and analytical characterization of this class of dendrons. With accessibility and analytical identification solved, we sought to evaluate both lower and higher generation dendrons for hepatocyte targeting as well as clearance of a model protein. We prepared a series of clearing agents where a single biotin is connected to glycodendrons displaying four, eight, sixteen or thirty-two α-thio-N-acetylgalactosamine (α-SGalNAc) units, resulting in compounds with molecular weights ranging from 2 to 17 kDa, respectively. These compounds were fully characterized by LCMS and NMR. We then evaluated the capacity of these agents to clear a model (131)I-labeled single chain variable fragment antibody-streptavidin ((131)I-scFv-SAv) fusion protein from blood and tissue in mice, and compared their clearing efficiencies to that of a 500 kDa dextran-biotin conjugate. Glycodendrons and dextran-biotin exhibited enhanced blood clearance of the scFv-SAv construct. Biodistribution analysis showed liver targeting/uptake of the scFv-SAv construct to be 2-fold higher for compounds 1 to 4, as well as for the 500 kDa dextran, over saline. Additionally, the data suggest the glycodendrons clear through the liver, whereas the dextran through reticuloendothelial system (RES) metabolism.


Asunto(s)
Acetilgalactosamina/metabolismo , Dendrímeros/metabolismo , Hígado/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Anticuerpos de Cadena Única/metabolismo , Acetilgalactosamina/farmacocinética , Animales , Dendrímeros/farmacocinética , Femenino , Radioisótopos de Yodo , Ratones , Proteínas Recombinantes de Fusión/sangre , Anticuerpos de Cadena Única/sangre , Estreptavidina/genética
14.
Nucleic Acid Ther ; 22(6): 380-90, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23181701

RESUMEN

Effective in vivo delivery of small interfering (siRNA) has been a major obstacle in the development of RNA interference therapeutics. One of the first attempts to overcome this obstacle utilized intravenous injection of cholesterol-conjugated siRNA (chol-siRNA). Although studies in mice revealed target gene knockdown in the liver, delivery was relatively inefficient, requiring 3 daily injections of 50 mg/kg of chol-siRNA to obtain measurable reduction in gene expression. Here we present a new delivery approach that increases the efficacy of the chol-siRNA over 500-fold and allows over 90% reduction in target gene expression in mice and, for the first time, high levels of gene knockdown in non-human primates. This improved efficacy is achieved by the co-injection of a hepatocyte-targeted and reversibly masked endosomolytic polymer. We show that knockdown is absolutely dependent on the presence of hepatocyte-targeting ligand on the polymer, the cognate hepatocyte receptor, and the cholesterol moiety of the siRNA. Importantly, we provide evidence that this increase in efficacy is not dependent on interactions between the chol-siRNA with the polymer prior to injection or in the bloodstream. The simplicity of the formulation and efficacy of this mode of siRNA delivery should prove beneficial in the use of siRNA as a therapeutic.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Colesterol/administración & dosificación , Endosomas/efectos de los fármacos , Polivinilos/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/farmacocinética , Animales , Apolipoproteínas B/sangre , Apolipoproteínas B/genética , Receptor de Asialoglicoproteína/genética , Receptor de Asialoglicoproteína/metabolismo , Colesterol/farmacocinética , Factor VII/genética , Factor VII/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Técnicas de Transferencia de Gen , Hepatocitos/metabolismo , Lípidos/sangre , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Polivinilos/farmacocinética , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacocinética , Receptores de LDL/genética , Receptores de LDL/metabolismo
15.
Biomaterials ; 32(17): 4118-29, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21429574

RESUMEN

There is an urgent need for novel polymeric carriers that can selectively deliver a large dose of chemotherapeutic agents into hepatic cancer cells to achieve high therapeutic activity with minimal systemic side effects. PAMAM dendrimers are characterized by a unique branching architecture and a large number of chemical surface groups suitable for coupling of chemotherapeutic agents. In this article, we report the coupling of N-acetylgalactosamine (NAcGal) to generation 5 (G5) of poly(amidoamine) (PAMAM-NH2) dendrimers via peptide and thiourea linkages to prepare NAcGal-targeted carriers used for targeted delivery of chemotherapeutic agents into hepatic cancer cells. We describe the uptake of NAcGal-targeted and non-targeted G5 dendrimers into hepatic cancer cells (HepG2) as a function of G5 concentration and incubation time. We examine the contribution of the asialoglycoprotein receptor (ASGPR) to the internalization of NAcGal-targeted dendrimers into hepatic cancer cells through a competitive inhibition assay. Our results show that uptake of NAcGal-targeted G5 dendrimers into hepatic cancer cells occurs via ASGPR-mediated endocytosis. Internalization of these targeted carriers increased with the increase in G5 concentration and incubation time following Michaelis-Menten kinetics characteristic of receptor-mediated endocytosis. These results collectively indicate that G5-NAcGal conjugates function as targeted carriers for selective delivery of chemotherapeutic agents into hepatic cancer cells.


Asunto(s)
Acetilgalactosamina/farmacocinética , Antineoplásicos/farmacología , Dendrímeros/síntesis química , Dendrímeros/farmacocinética , Portadores de Fármacos/síntesis química , Poliaminas/farmacología , Transporte Biológico , Sistemas de Liberación de Medicamentos/métodos , Endocitosis , Células Hep G2 , Humanos , Poliaminas/química , Polímeros
16.
Toxicol Appl Pharmacol ; 216(2): 303-8, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16842833

RESUMEN

Nutritional selenium compounds are transformed to the common intermediate selenide and then utilized for selenoprotein synthesis or excreted in urine mostly as 1beta-methylseleno-N-acetyl-Dd-galactosamine (selenosugar). Since the biological significance of selenosugar formation is unknown, we investigated their role in the formation of selenoenzymes in selenium deficiency. Rats were depleted of endogenous natural abundance selenium with a single stable isotope ((82)Se) and then made Se-deficient. (76)Se-Selenosugar was administered intravenously to the rats and their urine, serum, liver, kidneys and testes were subjected to speciation analysis with HPLC inductively coupled argon plasma mass spectrometry. Most (76)Se was recovered in its intact form (approximately 80% of dose) in urine within 1 h. Speciation analysis revealed that residual endogenous natural abundance selenium estimated by (77)Se and (78)Se was negligible and distinct distributions of the labeled (76)Se were detected in the body fluids and organs without interference from the endogenous natural abundance stable isotope. Namely, intact (76)Se-selenosugar was distributed to organs after the injection, and (76)Se was used for selenoprotein synthesis. Oxidation to methylseleninic acid and/or hydrolysis of the selenoacetal group to methylselenol were proposed to the transformation of selenosugar for the reuse. Effective use of an enriched stable isotope as an absolute label in hosts depleted of natural abundance isotopes was discussed for application in tracer experiments.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Compuestos de Organoselenio/farmacocinética , Selenio/deficiencia , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/farmacocinética , Animales , Biotransformación , Cromatografía Líquida de Alta Presión , Inyecciones Intravenosas , Isótopos , Masculino , Espectrometría de Masas , Compuestos de Organoselenio/administración & dosificación , Ratas , Ratas Wistar , Selenio/metabolismo , Selenoproteínas/análisis , Selenoproteínas/biosíntesis , Selenito de Sodio/farmacocinética , Selenito de Sodio/orina , Distribución Tisular
17.
Proc Natl Acad Sci U S A ; 102(47): 17125-9, 2005 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-16286643

RESUMEN

Endogenous ligands have not, to date, been identified for the asialoglycoprotein receptor (ASGP-R), which is abundantly expressed by parenchymal cells in the liver of mammals. On the basis of the rapid clearance of BSA bearing multiple chemically coupled sialic acid (Sia)alpha2,6GalNAcbeta1,4GlcNAcbeta1,2Man tetrasaccharides (SiaGGnM-BSA) from the circulation, and the ability of the ASGP-R hepatic lectin-1 subunit to bind SiaGGnM-BSA, we previously proposed that glycoproteins modified with structures terminating with Siaalpha2,6GalNAc may represent previously unrecognized examples of endogenous ligands for this receptor. Here, we have taken a genetic approach using wild-type and ASGP-R-deficient mice to determine that the ASGP-R in vivo does indeed account for the rapid clearance of glycoconjugates terminating with Siaalpha2,6GalNAc. We have also determined that the ASGP-R is able to bind core-substituted oligosaccharides with the terminal sequence Siaalpha2,6Galbeta1,4GlcNAc but not those with the terminal Siaalpha2,3Galbeta1,4GlcNAc. We propose that glycoproteins bearing terminals Siaalpha2,6GalNAc and Siaalpha2,6Gal are endogenous ligands for the ASGP-R, and that the ASGP-R helps to regulate the relative concentration of serum glycoproteins bearing alpha2,6-linked Sia.


Asunto(s)
Acetilgalactosamina/farmacocinética , Receptor de Asialoglicoproteína/sangre , Glicoconjugados/farmacocinética , Ácido N-Acetilneuramínico/farmacocinética , Acetilgalactosamina/sangre , Acetilgalactosamina/química , Animales , Receptor de Asialoglicoproteína/deficiencia , Receptor de Asialoglicoproteína/genética , Unión Competitiva/genética , Secuencia de Carbohidratos , Galactosa/sangre , Galactosa/farmacocinética , Glicoconjugados/sangre , Glicoconjugados/química , Ligandos , Tasa de Depuración Metabólica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Ácido N-Acetilneuramínico/sangre , Ácido N-Acetilneuramínico/química , Unión Proteica/genética , Estructura Terciaria de Proteína , Ratas , Especificidad de la Especie , Especificidad por Sustrato
18.
J Med Chem ; 47(23): 5798-808, 2004 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-15509179

RESUMEN

A novel glycolipid has been prepared that contains a cluster glycoside with an unusually high affinity for the asialoglycoprotein receptor (ASGPr) and a bile acid moiety that mediates stable incorporation into lipidic particles. The glycolipid spontaneously associated with low-density lipoproteins (LDL) and high-density lipoproteins (HDL) within human and murine plasma, and loading of lipoproteins with this glycolipid resulted in an efficient dose-dependent recognition and uptake of LDL and HDL by the liver (and not by spleen) upon intravenous injection into wild-type mice. Preinjection with asialoorosomucoid largely inhibited the uptake, establishing that both HDL and LDL were selectively recognized and processed by the ASGPr on liver parenchymal cells. Finally, repeated intravenous administration of the glycolipid to hyperlipidemic LDL receptor-deficient mice evoked an efficient and persistent cholesterol-lowering effect. These results indicate that the glycolipid may be a promising alternative for the treatment of hyperlipidemic patients who do not respond sufficiently to current cholesterol-lowering therapies.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Acetilgalactosamina/síntesis química , Receptor de Asialoglicoproteína/efectos de los fármacos , Glucolípidos/síntesis química , Acetilgalactosamina/farmacocinética , Acetilgalactosamina/farmacología , Animales , Anticolesterolemiantes/síntesis química , Anticolesterolemiantes/farmacocinética , Anticolesterolemiantes/farmacología , Receptor de Asialoglicoproteína/metabolismo , Proteínas Sanguíneas/metabolismo , Diseño de Fármacos , Glucolípidos/farmacocinética , Glucolípidos/farmacología , Humanos , Hiperlipidemias/tratamiento farmacológico , Hiperlipidemias/genética , Radioisótopos de Yodo , Marcaje Isotópico , Lipoproteínas HDL/sangre , Lipoproteínas HDL/química , Lipoproteínas LDL/sangre , Lipoproteínas LDL/química , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Receptores de LDL/genética
19.
Cancer Res ; 53(3): 658-62, 1993 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-8425200

RESUMEN

A method was elaborated for high-yield 125I-trap labeling of rat colon carcinoma cells using conjugates of dichlorotriazine aminofluorescein and bovine serum albumin substituted with either N-acetylgalactosamine or N-acetylglucosamine as vehicles. Fluorescence microscopy revealed that the ligands accumulated in perinuclear vesicles that were probably lysosomes. Monensin inhibited accumulation by 40%, signifying receptor-mediated endocytosis. Competition experiments revealed that the same receptor(s) mediated endocytosis of the two neoglycoproteins. Accumulation of label was greatly enhanced in the absence of serum, resulting in a labeling efficiency of at least 15 cpm/cell, with no sign of toxic effects. At least 75% of the initially accumulated radioactivity resided in the cells 4 days after labeling. After that the loss of radioactivity was linear with time and stabilized at 1.1%/day for at least 2 weeks. Injection of labeled carcinoma cells i.v. into syngeneic rats revealed a very rapid clearance from the circulation. Isolation of the liver cells 24 h later revealed that a great proportion of the administered cells or their remnants had been engulfed by sinusoidal Kupffer and endothelial cells; the parenchymal cells contained a smaller proportion of label. In conclusion, we have developed a technique of labeling colon carcinoma cells with 125I and fluorescein utilizing specific lectin-like receptors for endocytosis. Since the label is trapped intralysosomally, it will also label Kupffer cells and other members of the reticuloendothelial system after internalization. These features make the procedure well suited for studies on the fate of the colon carcinoma cells after administration in vivo. Since the label is trapped intralysosomally for an extended length of time, parameters such as the formation of metastasis and elimination by phagocytosis can readily be determined.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Acetilglucosamina/análogos & derivados , Adenocarcinoma/patología , Neoplasias del Colon/patología , Endocitosis/fisiología , Fluoresceína-5-Isotiocianato/farmacocinética , Fluoresceínas/farmacocinética , Células Neoplásicas Circulantes/patología , Albúmina Sérica Bovina/farmacocinética , Acetilgalactosamina/farmacocinética , Acetilgalactosamina/farmacología , Acetilglucosamina/farmacocinética , Acetilglucosamina/farmacología , Adenocarcinoma/sangre , Adenocarcinoma/metabolismo , Animales , Neoplasias del Colon/sangre , Neoplasias del Colon/metabolismo , Endocitosis/efectos de los fármacos , Fluoresceína-5-Isotiocianato/farmacología , Fluoresceínas/farmacología , Radioisótopos de Yodo , Hígado/citología , Masculino , Microscopía Fluorescente , Monensina/farmacología , Ratas , Ratas Endogámicas , Sensibilidad y Especificidad , Albúmina Sérica Bovina/farmacología
20.
Tohoku J Exp Med ; 168(2): 317-21, 1992 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1306316

RESUMEN

In order to evaluate the role of hexosamine metabolism in tumor tissue, we studied the biodistribution of N-(F-18)-fluoroacetyl-D-glucosamine (FAGlu) in male Donryu rats bearing poorly differentiated hepatomas (AH109A and AH272). Compare with the former result of the high tumor uptake of FAGlu in C3H/He mice bearing well differentiated spontaneous hepatoma, the tumor uptakes of FAGlu in these tumors showed the lower values. This suggested that spontaneous hepatoma maintained a high activity of glucosamine metabolism, while poorly differentiated hepatoma had little activity. Metabolism of glucosamine in tumor tissue may be another marker for characterizing tumors. We also discuss the tissue distribution of new F-18 labeled hexosamines, N-(F-18)-fluoroacetyl-D-mannosamine and N-(F-18)-fluoroacetyl-D-galactosamine in tumor bearing rats.


Asunto(s)
Acetilglucosamina/análogos & derivados , Radioisótopos de Flúor/farmacocinética , Neoplasias Hepáticas Experimentales/metabolismo , Acetilgalactosamina/análogos & derivados , Acetilgalactosamina/farmacocinética , Acetilglucosamina/farmacocinética , Animales , Hexosaminas/farmacocinética , Masculino , Manosa/análogos & derivados , Manosa/farmacocinética , Ratas , Ratas Endogámicas , Distribución Tisular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA