Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Pharmacol ; 60(3): 521-7, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11502883

RESUMEN

We recently synthesized a lipophilic prodrug of 9-(2-phosphonyl-methoxyethyl)adenine (PMEA), designated PMEA-LO, and incorporated it into reconstituted lactosylated high-density lipoprotein (LacNeoHDL). In a rat model, LacNeoHDL-associated PMEA-LO was internalized by the asialoglycoprotein receptor on parenchymal liver cells and converted into its active diphosphorylated metabolite. To further evaluate the therapeutic potential of the carrier-associated prodrug, we examined in this study the processing of (125)I-labeled PMEA-LO--loaded LacNeoHDL by HepG2 cells. Upon incubation with HepG2 cells, PMEA-LO--loaded LacNeoHDL became rapidly cell-associated. The association was saturable and of high-affinity (k(d) = 3.8 +/- 0.4 nM). Asialofetuin, an established ligand for the asialoglycoprotein receptor, inhibited the association by >75%, which confirms the role of the asialoglycoprotein receptor. Association of the prodrug-loaded particles to HepG2 cells was coupled to degradation. Radiolabeled degradation products appeared in the culture medium with a lag phase of 2 h. Asialofetuin and chloroquine inhibited secretion of degradation products by 75 to 80%, indicating that PMEA-LO--loaded LacNeoHDL is internalized via the asialoglycoprotein receptor and lysosomally processed. The therapeutic potential of LacNeoHDL-associated PMEA-LO was studied by measuring its effects on hepatitis B virus (HBV) replication in Hep AD38 cells (HBV-transfected HepG2 cells). LacNeoHDL-associated PMEA-LO effectively inhibited HBV DNA synthesis. The EC(50) value of carrier-associated PMEA-LO was 35 times lower than that of free PMEA (3.4 +/- 0.4 and 120 +/- 18 ng of PMEA/ml, respectively). We conclude that the present results, combined with our earlier in vivo disposition data, underscore the therapeutic potential and utility of PMEA-LO--loaded LacNeoHDL for treatment of chronic hepatitis B.


Asunto(s)
Adenina/farmacología , Antiinflamatorios no Esteroideos/farmacología , Sistemas de Liberación de Medicamentos , Virus de la Hepatitis B/efectos de los fármacos , Organofosfonatos , Adenina/administración & dosificación , Adenina/análogos & derivados , Antiinflamatorios no Esteroideos/administración & dosificación , Portadores de Fármacos , Virus de la Hepatitis B/fisiología , Humanos , Lactosa/metabolismo , Lipoproteínas HDL , Ácido Litocólico/química , Pruebas de Sensibilidad Microbiana , Células Tumorales Cultivadas , Replicación Viral/efectos de los fármacos
3.
Adv Drug Deliv Rev ; 47(2-3): 251-76, 2001 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-11311995

RESUMEN

Lipoproteins are endogenous particles that transport lipids through the blood to various cell types, where they are recognised and taken up via specific receptors. These particles are, therefore, excellent candidates for the targeted delivery of drugs to various tissues. For example, the remnant receptor and the asialoglycoprotein receptor (ASGPr), which are uniquely localised on hepatocytes, recognise chylomicrons and lactosylated high density lipopoteins (HDL), respectively. In addition, tumour cells of various origins overexpress the low density lipoprotein (LDL) receptor that recognises apolipoprotein E (apoE) on small triglyceride-rich particles and apoB-100 on LDL. Being endogenous, lipoproteins are biodegradable, do not trigger immune reactions, and are not recognised by the reticuloendothelial system (RES). However, their endogenous nature also hampers large-scale pharmaceutical application. In the past two decades, various research groups have successfully synthesised recombinant lipoproteins from commercially available natural and synthetic lipids and serum-derived or recombinant apolipoproteins, which closely mimic the metabolic behaviour of their native counterparts in animal models as well as humans. In this paper, we will summarise the studies that led to the development of these recombinant lipoproteins, and we will address the possibility of using these lipidic particles to selectively deliver a wide range of lipophilic, amphiphilic, and polyanionic compounds to hepatocytes and tumour cells. In addition, the intrinsic therapeutic activities of recombinant chylomicrons and HDL in sepsis and atherosclerosis will be discussed.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Hepatocitos/metabolismo , Lipoproteínas/administración & dosificación , Profármacos/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Animales , Arteriosclerosis/tratamiento farmacológico , Quilomicrones/administración & dosificación , Quilomicrones/química , Humanos , Lipoproteínas/química , Lipoproteínas HDL/administración & dosificación , Lipoproteínas HDL/química , Lipoproteínas LDL/administración & dosificación , Lipoproteínas LDL/química , Profármacos/química , Proteínas Recombinantes/química , Choque Séptico/tratamiento farmacológico , Tensoactivos/administración & dosificación , Tensoactivos/química , Células Tumorales Cultivadas/metabolismo
4.
FASEB J ; 14(12): 1784-92, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10973928

RESUMEN

The device of new hepatotrophic prodrugs of the antiviral nucleoside 9-(2-phosphonylmethoxyethyl)adenine (PMEA) with specificity for the asialoglycoprotein receptor on parenchymal liver cells is described. PMEA was conjugated to bi- and trivalent cluster glycosides (K(GN)(2) and K(2)(GN)(3), respectively) with nanomolar affinity for the asialoglycoprotein receptor. The liver uptake of the PMEA prodrugs was more than 10-fold higher than that of the parent drug (52+/-6% and 62+/-3% vs. 4.8+/-0.7% of the injected dose for PMEA) and could be attributed for 90% to parenchymal cells. Accumulation of the PMEA prodrugs in extrahepatic tissue (e.g., kidney, skin) was substantially reduced. The ratio of parenchymal liver cell-to-kidney uptake-a measure of the prodrugs therapeutic window-was increased from 0.058 +/- 0.01 for PMEA to 1.86 +/- 0.57 for K(GN)(2)-PMEA and even 2.69 +/- 0.24 for K(2)(GN)(3)-PMEA. Apparently both glycosides have a similar capacity to redirect (antiviral) drugs to the liver. After cellular uptake, both PMEA prodrugs were converted into the parent drug, PMEA, during acidification of the lysosomal milieu (t(1/2) approximately 100 min), and the released PMEA was rapidly translocated into the cytosol. The antiviral activity of the prodrugs in vitro was dramatically enhanced as compared to the parent drug (5- and 52-fold for K(GN)(2)-PMEA and K(2)(GN)(3)-PMEA, respectively). Given the 15-fold enhanced liver uptake of the prodrugs, we anticipate that the potency in vivo will be similarly increased. We conclude that PMEA prodrugs have been developed with greatly improved pharmacokinetics and therapeutic activity against viral infections that implicate the liver parenchyma (e.g., HBV). In addition, the significance of the above prodrug concept also extends to drugs that intervene in other liver disorders such as cholestasis and dyslipidemia.


Asunto(s)
Adenina/análogos & derivados , Adenina/farmacología , Antivirales/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Hígado/metabolismo , Organofosfonatos , Profármacos/farmacología , Adenina/sangre , Adenina/metabolismo , Adenina/farmacocinética , Animales , Antivirales/sangre , Antivirales/metabolismo , Antivirales/farmacocinética , Humanos , Masculino , Profármacos/metabolismo , Profármacos/farmacocinética , Ratas , Ratas Wistar , Tritio , Células Tumorales Cultivadas
5.
Nucleic Acids Res ; 28(14): 2717-25, 2000 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10908328

RESUMEN

Several studies have shown improved efficacy of cholesteryl-conjugated phosphorothioate antisense oligodeoxynucleotides. To gain insight into the mechanisms of the improved efficacy in vivo, we investigated the disposition of ISIS-9388, the 3'-cholesterol analog of the ICAM-1-specific phosphorothioate oligodeoxynucleotide ISIS-3082, in rats. Intravenously injected [(3)H]ISIS-9388 was cleared from the circulation with a half-life of 49.9 +/- 2.2 min (ISIS-3082, 23.3 +/- 3.8 min). At 3 h after injection, the liver contained 63.7 +/- 3. 3% of the dose. Compared to ISIS-3082, the hepatic uptake of ISIS-9388 is approximately 2-fold higher. Endothelial, Kupffer and parenchymal cells accounted for 45.7 +/- 5.7, 33.0 +/- 5.9 and 21.3 +/- 2.6% of the liver uptake of [(3)H]ISIS-9388, respectively, and intracellular concentrations of approximately 2, 75 and 50 microM, respectively, could be reached in these cells (1 mg/kg dose). Preinjection with polyinosinic acid or poly-adenylic acid reduced the hepatic uptake of [(3)H]ISIS-9388, which suggests the involvement of (multiple) scavenger receptors. Size exclusion chromatography of mixtures of the oligonucleotides and rat plasma indicated that ISIS-9388 binds to a larger extent to high molecular weight proteins than ISIS-3082. Analysis by agarose gel electrophoresis indicated that ISIS-9388 binds more tightly to plasma proteins than ISIS-3082. The different interaction of the oligonucleotides with plasma proteins possibly explains their different dispositions. We conclude that cholesterol conjugation results in high accumulation of phosphorothioate oligodeoxynucleotides in various liver cell types, which is likely to be beneficial for antisense therapy of liver-associated diseases.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Colesterol/análogos & derivados , Colesterol/farmacocinética , Hígado/metabolismo , Proteínas de la Membrana , Oligodesoxirribonucleótidos Antisentido/farmacocinética , Receptores de Lipoproteína , Tionucleótidos/farmacocinética , Animales , Colesterol/sangre , Colesterol/química , Hígado/citología , Masculino , Oligodesoxirribonucleótidos Antisentido/sangre , Oligodesoxirribonucleótidos Antisentido/química , Unión Proteica , Ratas , Ratas Wistar , Receptores Inmunológicos/metabolismo , Receptores Depuradores , Receptores Depuradores de Clase B , Tionucleótidos/sangre , Tionucleótidos/química , Tritio
6.
Antimicrob Agents Chemother ; 44(3): 477-83, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10681306

RESUMEN

Our aim is to selectively deliver 9-(2-phosphonylmethoxyethyl)adenine (PMEA) to parenchymal liver cells, the primary site of hepatitis B virus (HBV) infection. Selective delivery is necessary because PMEA, which is effective against HBV in vitro, is hardly taken up by the liver in vivo. Lactosylated reconstituted high-density lipoprotein (LacNeoHDL), a lipid particle that is specifically internalized by parenchymal liver cells via the asialoglycoprotein receptor, was used as the carrier. PMEA could be incorporated into the lipid moiety of LacNeoHDL by attaching, via an acid-labile bond, lithocholic acid-3alpha-oleate to the drug. The uptake of the lipophilic prodrug (PMEA-LO) by the liver was substantially increased after incorporation into LacNeoHDL. Thirty minutes after injection of [(3)H]PMEA-LO-loaded LacNeoHDL into rats, the liver contained 68.9% +/- 7.7% of the dose (free [(3)H]PMEA, <5%). Concomitantly, the uptake by the kidney was reduced to <2% of the dose (free [(3)H]PMEA, >45%). The hepatic uptake of PMEA-LO-loaded LacNeoHDL occurred mainly by parenchymal cells (88.5% +/- 8.2% of the hepatic uptake). Moreover, asialofetuin inhibited the liver association by >75%, indicating uptake via the asialoglycoprotein receptor. The acid-labile linkage in PMEA-LO, designed to release PMEA during lysosomal processing of the prodrug-loaded carrier, was stable at physiological pH but was hydrolyzed at lysosomal pH (half-life, 60 to 70 min). Finally, subcellular fractionation indicates that the released PMEA is translocated to the cytosol, where it is converted into its active diphosphorylated metabolite. In conclusion, lipophilic modification and incorporation of PMEA into LacNeoHDL improves the biological fate of the drug and may lead to an enhanced therapeutic efficacy against chronic hepatitis B.


Asunto(s)
Adenina/análogos & derivados , Antivirales/administración & dosificación , Antivirales/farmacocinética , Hepatitis B/tratamiento farmacológico , Hígado/metabolismo , Organofosfonatos , Adenina/administración & dosificación , Adenina/sangre , Adenina/química , Adenina/farmacocinética , Animales , Antivirales/sangre , Antivirales/química , Cromatografía/métodos , Portadores de Fármacos , Lipoproteínas HDL/metabolismo , Ácido Litocólico/análogos & derivados , Ácido Litocólico/síntesis química , Ácido Litocólico/metabolismo , Ácido Litocólico/farmacología , Hígado/citología , Hígado/efectos de los fármacos , Masculino , Profármacos/administración & dosificación , Profármacos/síntesis química , Profármacos/farmacocinética , Ratas , Ratas Wistar , Distribución Tisular , Tritio
7.
Biochem Pharmacol ; 59(11): 1407-16, 2000 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10751550

RESUMEN

Low-density lipoprotein (LDL) has been proposed as carrier for the selective delivery of anticancer drugs to tumor cells. We reported earlier the association of several lipidic steroid-conjugated anticancer oligodeoxynucleotides (ODNs) with LDL. In the present study, we determined the stability of these complexes. When the complexes were incubated with a mixture of high-density lipoprotein and albumin, or with rat plasma, the oleoyl steroid-conjugated ODNs appeared to be more stably associated with LDL than the cholesteryl-conjugated ODN. Intravenously injected free lipid-ODNs were very rapidly cleared from the circulation of rats. The area under the curve (AUC) of the lipid-ODNs in plasma was <0.4 microg x min/mL. After complexation with LDL, plasma clearance of the lipid-ODNs was delayed. This was most evident for ODN-5, the ODN conjugated with the oleoyl ester of lithocholic acid (AUC = 6.82 +/- 1.34 microg x min/mL). The AUC of ODN-4, a cholesteryl-conjugated ODN, was 1.49 +/- 0.37 microg x min/mL. In addition, the liver uptake of the LDL-complexed lipid-ODNs was reduced. The lipid-ODNs were also administered as a complex with lactosylated LDL, a modified LDL particle that is selectively taken up by the liver. A high proportion of ODN-5 was transported to the liver along with lactosylated LDL (69.1 +/- 8.1% of the dose at 15 min after injection), whereas much less ODN-4 was transported (36.6 +/- 0.1% of the dose at 15 min after injection). We conclude that the oleoyl ester of lithocholic acid is a more potent lipid anchor than the other steroid lipid anchors. Because of the stable association, the oleoyl ester of lithocholic acid is an interesting candidate for tumor targeting of anticancer ODNs with lipoproteins.


Asunto(s)
Antineoplásicos/farmacocinética , Lipoproteínas LDL/farmacocinética , Hígado/metabolismo , Oligodesoxirribonucleótidos Antisentido/farmacocinética , Esteroides/farmacocinética , Animales , Antineoplásicos/sangre , Humanos , Lactosa/metabolismo , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/sangre , Masculino , Tasa de Depuración Metabólica , Oligodesoxirribonucleótidos Antisentido/sangre , Ratas , Ratas Wistar , Albúmina Sérica/metabolismo , Esteroides/sangre
8.
Pharm Res ; 16(8): 1179-85, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10468017

RESUMEN

PURPOSE: 9-(2-Phosphonylmethoxyethyl)adenine (PMEA), a potent inhibitor of Hepatitis B virus replication, is in vivo hardly taken up by parenchymal liver cells (the site of infection). Our aim is to examine whether lactosylated reconstituted HDL (LacNeoHDL), a lipidic particle that is specifically internalized by parenchymal liver cells, is a suitable carrier for the selective delivery of PMEA to this cell type. METHODS: To incorporate PMEA into LacNeoHDL, we synthesized a lipophilic prodrug (PMEA-LO) by coupling PMEA via an acid-labile phosphonamidate bond to lithocholic acid-3alpha-oleate. RESULTS: The yield of the synthesis was 52% ([3H]PMEA-LO: 24%). [3H]PMEA-LO readily incorporated into LacNeoHDL (13 molecules/particle) without affecting the size and net negative charge of the carrier. Further, incubation studies at lysosomal pH showed [3H]PMEA was completely released from the carrier whereas, at neutral pH or in plasma, appreciable release was not observed. CONCLUSIONS: The conjugation of PMEA with lithocholic acid-3alpha-oleate results in a lipophilic prodrug that readily associates with Lac-NeoHDL. The association of the prodrug does not affect the physicochemical properties of the particle, and PMEA is released from the carrier at lysosomal pH. These findings indicate that by using the prodrug approach, LacNeoHDL is a suitable carrier to deliver PMEA to parenchymal liver cells.


Asunto(s)
Adenina/análogos & derivados , Antivirales/administración & dosificación , Ácido Litocólico/análogos & derivados , Ácido Litocólico/síntesis química , Hígado/metabolismo , Organofosfonatos , Profármacos/síntesis química , Adenina/administración & dosificación , Adenina/farmacología , Animales , Antivirales/farmacología , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Humanos , Lipoproteínas HDL/metabolismo , Ácido Litocólico/metabolismo , Ácido Litocólico/farmacología , Hígado/efectos de los fármacos , Profármacos/metabolismo , Profármacos/farmacología , Ratas , Tritio
9.
J Pharmacol Exp Ther ; 286(3): 1166-70, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9732374

RESUMEN

It has been reported that conjugating acyclovir, a potent antiviral with low oral bioavailability, to L-valine increases its urinary excretion in rats. However, it was also reported that this increase is not found for the D-valine ester, suggesting that a carrier-mediated mechanism is involved in its intestinal absorption. Therefore, mechanisms involved in the transepithelial transport of L-valine-acyclovir were investigated using the intestinal cell line, Caco-2, as a model system for the intestinal epithelium. Only the mucosal-to-serosal transport of acyclovir was increased by conjugation with L-valine (approximately 7-fold), suggesting the involvement of a carrier-mediated mechanism. This conclusion was supported by the finding that this increase was saturable. The mucosal-to-serosal transport of L-valine-acyclovir could be inhibited by L-glycylsarcosine, but not by L-valine, suggesting the involvement of the dipeptide carrier. Also it was found that L-valine-acyclovir inhibits the uptake of cephalexin, a substrate for the oligopeptide transporter. Stability of the esters in either the mucosal or serosal bathing solution is more than 90% after completion of the transport study. However, after transport, the receiver solution contained approximately 90% of acyclovir. Based on these findings it was concluded that absorption of the L-valine ester of acyclovir occurs as a result of uptake by the oligopeptide transporter at the apical cell membrane followed by intracellular hydrolysis of the ester and efflux of acyclovir.


Asunto(s)
Aciclovir/farmacocinética , Antivirales/farmacocinética , Proteínas Portadoras/fisiología , Mucosa Intestinal/metabolismo , Valina/farmacocinética , Aciclovir/química , Transporte Biológico , Células CACO-2 , Cefalexina/farmacocinética , Relación Dosis-Respuesta a Droga , Estabilidad de Medicamentos , Humanos , Concentración de Iones de Hidrógeno
10.
Pharm Res ; 15(8): 1154-9, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9706043

RESUMEN

PURPOSE: General use of nucleoside analogues in the treatment of viral infections and cancer is often limited by poor oral absorption. Valacyclovir, a water soluble amino acid ester prodrug of acyclovir has been reported to increase the oral bioavailability of acyclovir but its absorption mechanism is unknown. This study characterized the intestinal absorption mechanism of 5' -amino acid ester prodrugs of the antiviral drugs and examined the potential of amino acid esters as an effective strategy for improving oral drug absorption. METHODS: Acyclovir (ACV) and Zidovudine (AZT) were selected as the different sugar-modified nucleoside antiviral agents and synthesized to L-valyl esters of ACV and AZT (L-Val-ACV and L-Val-AZT), D-valyl ester of ACV (D-Val-ACV) and glycly ester of ACV (Gly-ACV). The intestinal absorption mechanism of these 5' -amino acid ester prodrugs was characterized in three different experimental systems; in situ rat perfusion model, CHO/hPEPT1 cells and Caco-2 cells. RESULTS: Testing 5' -amino acid ester prodrugs of acyclovir and AZT, we found that the prodrugs increased the intestinal permeability of the parent nucleoside analogue 3- to 10-fold. The dose- dependent permeation enhancement was selective for L-amino acid esters. Competitive inhibition studies in rats and in CHO cells transfected with the human peptide transporter, hPEPT1, demonstrated that membrane transport of the prodrugs was mediated predominantly by the PEPT1 H+/dipeptide cotransporter even though these prodrugs did not possess a peptide bond. Finally, transport studies in Caco-2 cells confirmed that the 5' - amino acid ester prodrugs enhanced the transcellular transport of the parent drug. CONCLUSIONS: This study demonstrates that L-amino acid-nucleoside chimeras can serve as prodrugs to enhance intestinal absorption via the PEPT1 transporter, providing a novel strategy for improving oral therapy of nucleoside drugs.


Asunto(s)
Aciclovir/metabolismo , Antivirales/metabolismo , Proteínas Portadoras/metabolismo , Mucosa Intestinal/metabolismo , Simportadores , Zidovudina/metabolismo , Aciclovir/química , Aciclovir/farmacología , Animales , Antivirales/química , Antivirales/farmacología , Disponibilidad Biológica , Células CHO , Células CACO-2 , Cricetinae , Ésteres , Humanos , Absorción Intestinal , Transportador de Péptidos 1 , Ratas , Zidovudina/química , Zidovudina/farmacología
11.
Bioconjug Chem ; 9(3): 341-9, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9576808

RESUMEN

The high expression level of receptors for low-density lipoprotein (LDL) on tumor cells makes LDL an attractive carrier for selective delivery of drugs to these cells. The aim of this study is to allow incorporation of oncogene-directed antisense oligodeoxynucleotides (ODNs) into the lipid moiety of LDL. Therefore, ODNs were conjugated with oleic acid, cholesterol, and several other steroid lipids. These latter steroid lipids were synthesized starting from bile acids and were varied in lipophilicity by attaching oleic acid ester structures. The lipid structures, activated as pentafluorophenyl esters, were conjugated in solution phase to 3'-amino-tailed ODNs. The ODNs conjugated with lithocholic acid, oleic acid, and cholesterol could easily be purified by reversed phase (RP)-HPLC. However, the ODNs conjugated with the oleoyl steroid ester structures irreversibly bound to the column material. These highly lipidic ODNs were separated from the nonconjugated ODN by electrophoresis in a 1% low-melting agarose gel containing 0.1% Tween 20. This method was found to be very effective in isolating the ODNs conjugated to the oleoyl steroid ester structures. The ODNs conjugated with cholesterol and the oleoyl esters of lithocholic and cholenic acid associated readily and nearly completely with LDL. However, the less lipidic ODNs and the ODN conjugated with the dioleoyl ester of chenodeoxycholic acid did not and did incompletely associate, respectively. Lithocholic acid and oleic acid are probably not sufficiently lipophilic to induce association with LDL, whereas the dioleoyl ester structure is probably too bulky and extended to allow partitioning into the lipid moiety of LDL. We conclude that several of the lipid-ODNs can associate readily with LDL, enabling delivery of oncogene-directed antisense ODNs via the LDL receptor pathway.


Asunto(s)
Lípidos/química , Lipoproteínas LDL/metabolismo , Oligodesoxirribonucleótidos/química , Ácidos y Sales Biliares/química , Colesterol/análogos & derivados , Electroforesis en Gel de Agar , Estructura Molecular , Ácidos Oléicos/química , Oligonucleótidos Antisentido/genética , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas c-myb , Esteroides/química , Transactivadores/química
12.
Nucleic Acids Res ; 25(16): 3290-6, 1997 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-9241243

RESUMEN

Systemically administered phosphorothioate antisense oligodeoxynucleotides can specifically affect the expression of their target genes, which affords an exciting new strategy for therapeutic intervention. Earlier studies point to a major role of the liver in the disposition of these oligonucleotides. The aim of the present study was to identify the cell type(s) responsible for the liver uptake of phosphorothioate oligodeoxynucleotides and to examine the mechanisms involved. In our study we used ISIS-3082, a phosphorothioate antisense oligodeoxynucleotide specific for murine ICAM-1. Intravenously injected [3H]ISIS-3082 (dose: 1 mg/kg) was cleared from the circulation of rats with a half-life of 23.3+/-3.8 min. At 90 min after injection (>90% of [3H]ISIS-3082 cleared), the liver contained the most radioactivity, whereas the second-highest amount was recovered in the kidneys (40.5+/-1.4% and 17.9+/-1.3% of the dose, respectively). Of the remaining tissues, only spleen and bone marrow actively accumulated [3H]ISIS-3082. By injecting different doses of [3H]ISIS-3082, it was found that uptake by liver, spleen, bone marrow, and kidneys is saturable, which points to a receptor-mediated process. Subcellular fractionation of the liver indicates that ISIS-3082 is internalized and delivered to the lysosomes. Liver uptake occurs mainly (for 56.1+/-3.0%) by endothelial cells, whereas parenchymal and Kupffer cells account for 39.6+/-4.5 and 4.3+/-1.7% of the total liver uptake, respectively. Preinjection of polyinosinic acid substantially reduced uptake by liver and bone marrow, whereas polyadenylic acid was ineffective, which indicates that in these tissues scavenger receptors are involved in uptake. Polyadenylic acid, but not polyinosinic acid, reduced uptake by kidneys, which suggests renal uptake by scavenger receptors different from those in the liver. We conclude that scavenger receptors on rat liver endothelial cells play a predominant role in the plasma clearance of ISIS-3082. As scavenger receptors are also expressed on human endothelial liver cells, our findings are probably highly relevant for the therapeutic application of phosphorothioate oligodeoxynucleotides in humans. If the target gene is not localized in endothelial liver cells, the therapeutic effectiveness might be improved by developing delivery strategies that redirect the oligonucleotides to the actual target cells.


Asunto(s)
Hígado/metabolismo , Proteínas de la Membrana , Oligonucleótidos Antisentido/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Lipoproteína , Tionucleótidos/metabolismo , Animales , Endotelio/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Masculino , Tasa de Depuración Metabólica , Ratones , Polielectrolitos , Polímeros/metabolismo , Ratas , Ratas Wistar , Receptores Depuradores , Receptores Depuradores de Clase B , Fracciones Subcelulares/metabolismo , Distribución Tisular
13.
Clin Pharmacokinet ; 31(2): 131-55, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8853935

RESUMEN

The hepatitis B virus (HBV) is the world's most important chronic virus infection. The immunomodulator interferon-alpha (IFN alpha) is the only clinically applied drug available, despite its low response rate (approximately 30%) even in highly selected chronic carriers. Antiviral nucleoside analogues have proven to be potent inhibitors of viral replication in vitro, but their significant adverse effects which are, at least partially, due to their nonspecific body distribution, have forced the cessation of their clinical development in the past. For example, vidarabine causes severe neuromuscular toxicity, and fialuridine has caused fatal cases of liver and kidney failure in a recent clinical trial. Furthermore, the potential clinical application of (modified) antisense oligodeoxynucleotides, which are very specific inhibitors of viral replication, is hampered by their nonspecific body distribution, instability in serum and poor cell penetration. As infection and replication of HBV mainly occur in liver parenchymal cells, selective targeting of antiviral nucleoside analogues as well as antisense oligodeoxynucleotides to the liver would theoretically improve therapeutic efficacy. At present, conjugates of vidarabine and neoglycoproteins have entered clinical trials, and initial data suggest that therapeutic concentrations are achieved at lower dosages with minor adverse effects. These data have stimulated preclinical research on other liver-specific drug carriers for the selective delivery of HBV-active drugs such as glycosylated polymers and neolipoproteins: these approaches are outlined in this paper.


Asunto(s)
Antivirales/administración & dosificación , Hepatitis B/tratamiento farmacológico , Profármacos/administración & dosificación , Antivirales/efectos adversos , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Humanos , Vidarabina/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...