Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
3.
J Pharmacol Exp Ther ; 301(3): 1139-43, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12023548

ABSTRACT

We have synthesized a prodrug of the angiotensin-converting enzyme (ACE) inhibitor captopril by coupling this drug covalently to the low molecular weight protein (LMWP) lysozyme. Such drug-LMWP conjugates can be used for renal drug delivery, since LMWPs accumulate specifically in the proximal tubular cells of the kidney. In the present study, we compared the effects of captopril-lysozyme and free captopril in male Wistar rats. ACE activity in plasma and the kidney was measured after intravenous bolus injection of either the captopril-lysozyme conjugate (33 mg. kg(-1), corresponding to 0.2 mg. kg(-1) captopril) or equivalent dosages of free captopril and lysozyme. The administration of the captopril-lysozyme conjugate resulted in less plasma ACE inhibition and a longer-lasting renal ACE inhibition compared with the free drug. Effects on blood pressure and natriuresis were studied during intravenous infusion of captopril-lysozyme (275 mg. kg(-1). 6 h(-1) conjugate, corresponding to 5 mg. kg(-1). 6 h(-1) captopril) or an equimolar dosage of free captopril. Captopril-lysozyme did not affect systemic blood pressure, whereas free captopril lowered blood pressure significantly (-23 +/- 32% versus control after 6 h). Captopril-lysozyme increased natriuresis about 3-fold compared with control levels (260 +/- 32% after 6 h), whereas free captopril treatment resulted in a reduced sodium excretion (26 +/- 12%). Furthermore, captopril at a lower dose, which only moderately lowered blood pressure, showed an increased sodium excretion. We conclude that renal delivery of captopril using captopril-lysozyme results in reduced systemic activity and increased kidney-specific activity of the targeted drug.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Blood Pressure/drug effects , Captopril/administration & dosage , Drug Delivery Systems , Kidney/drug effects , Kidney/enzymology , Animals , Blood Pressure/physiology , Drug Delivery Systems/methods , Drug Delivery Systems/statistics & numerical data , Enzyme Activation/drug effects , Enzyme Activation/physiology , Infusions, Intravenous , Male , Muramidase/administration & dosage , Prodrugs/administration & dosage , Rats , Rats, Wistar
4.
Ren Fail ; 23(3-4): 397-409, 2001.
Article in English | MEDLINE | ID: mdl-11499555

ABSTRACT

Selective targeting of drugs to the kidney may enable an increased renal effectiveness combined with a reduction of extrarenal toxicity. Intrarenal delivery to the proximal tubular cell can be achieved using low-molecular-weight proteins, such as lysozyme. Administration of high dosages of lysozyme, required to study the effects of such conjugates in vivo, however, is restricted since a partial escape of the renal reabsorption and the occurrence of unwanted effects on systemic blood pressure and renal function may occur. The purpose of this study was to investigate the optimal parenteral administration schedule and the maximum dose of lysozyme, providing the most optimal tubular reabsorption and at the same time a minimal effect on blood pressure and renal hemodynamics, comparing continuous infusion of lysozyme with single dose injections. Urinary lysozyme excretion increased dose-dependently, both during continuous infusion and intravenous bolus injections. However, this loss of intact lysozyme into the urine was much higher after 3 injections of in total 250 mg x kg(-1) x 6 h(-1) (51.8+/-3.7% of the dose) compared to the same dose administered by continuous infusion (11.7+/-2.4%, P < 0.001). Continuous infusion of lysozyme up to 1000 mg x kg(-1) in 6 hours had no effect on systemic blood pressure, whereas a bolus injection of lysozyme (167 mg x kg(-1)) resulted in reversible blood pressure lowering of 52.2+/-2.2% (P<0.001). A dose-dependent decline of the glomerular filtration rate was observed at dosages of lysozyme higher than 100 mg x kg(-1) x 6 h(-1), with a maximal reduction of 53.0+/-3.7% after infusion of 1000 mg x kg(-1) x 6 h(-1). Effective renal plasma flow was less affected and only lowered statistically significant at dosages of 500 (-12.6+/-3.3%, P<0.05) to 1000 mg x kg(-1) x 6 h(-1) (-17.2+/-3.9%, P<0.01). We conclude that bolus injections of lysozyme should not be used for renal targeting purposes since it results in considerable tubular loss of lysozyme in the urine as well as cardiovascular side effects. In contrast, continuous infusion of lysozyme using dosages sufficient for renal drug targeting (maximally 15 mg x kg(-1) x h(-1)) only has minimal effects on blood pressure and renal hemodynamics, with a minimal urinary lysozyme loss as well.


Subject(s)
Anti-Infective Agents/administration & dosage , Blood Pressure/drug effects , Muramidase/administration & dosage , Renal Circulation/drug effects , Animals , Drug Carriers , Infusions, Parenteral , Kidney Diseases/drug therapy , Male , Maximum Tolerated Dose , Muramidase/urine , Rats , Rats, Wistar
5.
Eur J Clin Pharmacol ; 56(3): 219-23, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10952476

ABSTRACT

OBJECTIVE: To compare the efficacy, safety and pharmacokinetics of a newly developed controlled-release suppository (MSR) with MS Contin tablets (MSC) in cancer patients with pain. METHODS: In a double-blind, randomised, two-way cross-over trial, 25 patients with cancer pain were selected with a morphine (M) demand of 30 mg every 12 h. Patients were divided into two groups. Group 1 received active MSC (30 mg) and placebo MSR, followed by placebo MSC and active MSR (30 mg) each for a period of 5 days. Group 2 started with active MSR and placebo MSC, followed by active MSC and placebo MSR, each for a period of 5 days. Blood for determination of plasma concentration of morphine (M) and its 3- and 6-glucuronides (M3G, M6G) was collected, and area under the plasma concentration-time curve (AUC)0-12 h, peak plasma concentration (Cmax), time to reach Cmax (tmax), and CO and C12 of M, M6G and M3G were determined on day 5 and day 10. Intensity of pain experienced by each patient was assessed every 2 h on a 0-10 scale, while side effects and rescue medication were recorded. RESULTS: Twenty patients (ten patients in each group) completed the study. A pronounced inter-patient variability in plasma concentrations of M, M3G and M6G was observed after administration of both forms. Apart from the C0 and C12, no significant differences in AUC0-12 h, tmax and Cmax of morphine between the rectal and oral route of administration were found. In the case of the metabolites, it was found that AUC0-12 h and Cmax of M6G, and AUC0-12 h, Cmax, C0 and C12 of M3G after rectal administration were significantly lower than after oral administration. However, apart from the tmax of M6G, none of the pharmacokinetic parameters of M, M6G or M3G met the criteria for bioequivalence. There were no significant (P = 0.44) differences in pain intensity score between the oral and rectal forms within the two groups, regardless of the treatment sequence. No treatment differences in nausea, sedation or the demand on escape medication (acetaminophen tablets) between the rectal and oral forms were observed. CONCLUSION: The newly developed controlled-release M suppository is safe and effective and may be a useful alternative for oral morphine administration in patients with cancer pain.


Subject(s)
Analgesics, Opioid/administration & dosage , Morphine/administration & dosage , Neoplasms/physiopathology , Pain, Intractable/drug therapy , Adult , Aged , Aged, 80 and over , Biological Availability , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Morphine/adverse effects , Morphine/pharmacokinetics , Morphine Derivatives/blood , Suppositories
7.
J Pharmacol Exp Ther ; 288(1): 281-5, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9862782

ABSTRACT

Low-molecular-weight proteins (LMWPs) accumulate in the proximal tubular cells of the kidney, which makes these proteins interesting tools for renal drug targeting. We studied this approach using the LMWP lysozyme as a carrier for the angiotensin-converting enzyme inhibitor captopril. Captopril was conjugated to lysozyme via a disulfide bond. The pharmacokinetics of the captopril-lysozyme conjugate was studied in the rat. Only intact conjugate could be detected in the circulation. The total amount of captopril disulfides in the kidney was six times higher after administration of the conjugate than after the administration of an equivalent amount of free captopril. The conjugate was recovered in the urine partially as intact conjugate and partially as low-molecular-weight disulfides. The excretion of conjugate in the urine was not a consequence of the coupling of captopril to lysozyme because an equivalent bolus dose of native lysozyme was similarly excreted into the urine. By determination of the renal angiotensin-converting enzyme activity, we showed that the conjugate was degraded to the pharmacologically active captopril in vivo. We conclude that the coupling of captopril to the LMWP lysozyme results in increased captopril concentrations in the kidney and reduced captopril concentrations in the circulation.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Captopril/pharmacokinetics , Drug Delivery Systems , Kidney/metabolism , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Captopril/administration & dosage , Kidney/drug effects , Male , Metabolic Clearance Rate , Muramidase/administration & dosage , Muramidase/pharmacokinetics , Prodrugs/metabolism , Rats , Rats, Wistar , Time Factors
8.
Clin Sci (Lond) ; 95(5): 603-9, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9791047

ABSTRACT

1. Renal specific targeting of the non-steroidal anti-inflammatory drug naproxen was obtained by coupling to the low-molecular-mass protein lysozyme. A previous study showed that conjugation to lysozyme resulted in a 70-fold increase of naproxen accumulation in the kidney with a subsequent renal release of the active metabolite naproxen-lysine.2. In the present study we questioned whether naproxen-lysozyme is active in the rat kidney, inhibiting the urinary excretion of prostaglandin E2 and renal sodium and water excretion in salt-restricted baseline conditions as well as during frusemide treatment.3.A high dose of free naproxen (10 mg.day-1. kg-1) did not affect prostaglandin E2 excretion in baseline conditions (naproxen, 11+/-1 ng/8 h; vehicle, 13+/-4 ng/8 h), whereas sodium and water excretion were, respectively, 3.0 and 1.6 times lower in the naproxen group (P<0.05). Naproxen completely prevented the frusemide-induced increase (3-fold) in prostaglandin E2 excretion (naproxen 6.6+/-1.1 ng/8 h, vehicle 40+/-12 ng/8 h, P<0. 005). Frusemide-stimulated natriuresis and diuresis were, respectively, 1.6 (P<0.05) and 1.8 times (P<0.005) lower in the naproxen group.4.A dose of 2 mg.day-1.kg-1 lysozyme-conjugated naproxen did not affect prostaglandin E2 excretion in baseline conditions (conjugate, 18+/-2 ng/8 h; vehicle, 24+/-5 ng/8 h). The conjugate also had no effect on sodium and water excretion. However, the naproxen conjugate completely prevented the frusemide-induced increase (2-fold) in prostaglandin E2 excretion (conjugate, 16+/-3 ng/8 h; vehicle, 48+/-13 ng/8 h, P<0.05). Surprisingly, frusemide-induced natriuresis and diuresis were not affected by the conjugate.5. In conclusion, a renal specific delivery of the non-steroidal anti-inflammatory drug naproxen using lysozyme results in an inhibitory effect on renal prostaglandin E2 synthesis but does not affect the excretion of sodium and water, in contrast to free naproxen.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dinoprostone/biosynthesis , Kidney/drug effects , Naproxen/pharmacology , Animals , Dinoprostone/urine , Diuretics/pharmacology , Drug Carriers , Furosemide/pharmacology , Kidney/metabolism , Male , Muramidase , Natriuresis/drug effects , Rats , Rats, Wistar , Urination/drug effects
9.
Ren Fail ; 20(2): 211-7, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9574445

ABSTRACT

The low molecular weight protein (LMWP) lysozyme is a suitable drug carrier for renal drug targeting. When the tubular reabsorption of a LMWP can be prevented, the protein will be excreted in the urine. In this way, lysozyme (LZM) conjugates might also be used as carriers for targeting to the urinary tract. Since positive domains on the protein surface are important for the interaction with the tubular uptake-receptor, we studied the urinary excretion of a drug-LZM conjugate with and without positive charge on the LMWP. We synthesized two conjugates with the fluorescent compound fluorescein. A positively charged conjugate was obtained by reacting fluorescein isothiocyanate (FITC) with LZM at a 1:1 molar to molar ratio; this conjugate contained six free primary aminogroups. The conjugate without positively charged groups was obtained by reacting the remaining free primary aminogroups of the FITC-LZM with succinic anhydride (Suc). The Suc-FITC-LZM contained only 0.2 free primary aminogroups per molecule. We studied the pharmacokinetics of the conjugates in freely moving Wistar rats. The FITC-LZM conjugate was excreted intactly into the urine for 29 +/- 4% of the injected dose. The Suc-FITC-LZM was excreted into the urine intactly for 45 +/- 4%. These data indicate that the excretion of a drug-LMWP conjugate into the urine can be increased by decreasing the positive charge on the carrier surface. Such a carrier may be an attractive candidate for drug targeting to the bladder.


Subject(s)
Anti-Infective Agents/administration & dosage , Drug Delivery Systems , Kidney/drug effects , Muramidase/administration & dosage , Urinary Bladder/drug effects , Animals , Anti-Infective Agents/pharmacokinetics , Anti-Infective Agents/urine , Drug Carriers , Fluorescein-5-isothiocyanate/administration & dosage , Fluorescein-5-isothiocyanate/pharmacokinetics , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/pharmacokinetics , Infusions, Intravenous , Kidney/metabolism , Male , Muramidase/pharmacokinetics , Muramidase/urine , Rats , Rats, Wistar , Succinic Anhydrides/administration & dosage , Succinic Anhydrides/pharmacokinetics , Urinary Bladder/metabolism
10.
J Pharmacol Toxicol Methods ; 38(1): 47-51, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9339416

ABSTRACT

Studies on short-term renal responses to (pharmacological) intervention require accurate and multiple collection of urine samples. Several invasive techniques have been described for frequent urine collection of the conscious rat, each having their own limitations. No data are available about the feasibility of the spontaneously voiding, freely moving rat for this purpose. In the present study, bladder voidings of six rats were time-registered and collected separately for several days. The data show a considerable 24-h variation coefficient of both the voided volume and the bladder collection time with a poor correlation between the two parameters. Forced diuresis induced by continuous i.v. infusion (2 ml/h) increased the frequency of urine voiding and thus the time-resolution of the urine-production pattern. However, this method failed to reduce the variation coefficient of the voided volume, the collection time, and the correlation between the two parameters. The fact that variations in creatinine excretion paralleled the variation in urinary flow suggests that both phenomena are likely be due to incomplete bladder emptying. Correction for this incomplete bladder collection, using the creatinine excretion, indeed reduced the variation coefficient of sodium excretion successfully from 61 +/- 17% to 29 +/- 5% during normal diuresis and from 56 +/- 19% to 22 +/- 6% during forced diuresis. In conclusion, the spontaneously voiding, freely moving rat can be used for short-term renal response studies if the collected urine samples are corrected for incomplete bladder emptying using urinary creatinine concentrations. This procedure allows the detection of changes in a urinary parameter if this exceeds a 40% deviation of the normal value.


Subject(s)
Kidney/physiology , Specimen Handling/instrumentation , Urine , Animals , Consciousness , Creatinine/metabolism , Diuresis/drug effects , Diuresis/physiology , Dopamine/pharmacology , Equipment Design , Kidney/drug effects , Male , Rats , Rats, Wistar , Reproducibility of Results , Sodium/metabolism , Urination/drug effects , Urination/physiology
11.
J Chromatogr B Biomed Sci Appl ; 693(2): 484-8, 1997 Jun 06.
Article in English | MEDLINE | ID: mdl-9210456

ABSTRACT

One possibility to optimize the therapeutic application of dopaminergic compounds with a catechol function is the reversible protection of this moiety using a prodrug approach. Important features in this respect are a proper chemical stability in the gastrointestinal tract, an adequate release rate after arrival in the blood stream or the possibility to cross the blood-brain barrier. A HPLC method was developed to measure the hydrolysis of prodrugs of dopamine and epinine directly. The method is based on reversed-phase separation followed by post-column ion-pair extraction with a fluorescent counter-ion. The separation of di-isobutyryl esters of dopamine and epinine is obtained within 10 min while the more hydrophobic dopaminergic esters, di-benzoyl and di-pivaloyl dopamine, are retained for 30 min. The precision of the assay measuring 160 ng dibudop and 100 ng ibopamine was 1.2 and 1.0%, respectively. The detection limit of all prodrugs tested was approximately 10 ng.


Subject(s)
Dopamine Agents/analysis , Prodrugs/analysis , Chromatography, High Pressure Liquid , Dopamine Agents/blood , Dopamine Agents/pharmacokinetics , Humans , Hydrolysis , Prodrugs/pharmacokinetics , Spectrometry, Fluorescence
12.
J Chromatogr B Biomed Sci Appl ; 693(1): 181-9, 1997 May 23.
Article in English | MEDLINE | ID: mdl-9200533

ABSTRACT

This study describes the development and comparison of two HPLC methods for the analysis of the antihypertensive drug captopril. The first method is based on a precolumn derivatization of captopril with the fluorescent label monobromobimane (MBB). The second method is based on a postcolumn reaction with the fluorescent reagent o-phthaldialdehyde (OPA). Since the disulfide metabolites of captopril can be reconverted to the active drug in vivo, the bioanalysis of captopril should involve both the determination of its free thiol form (free captopril) and the total amount of free thiol and reducible disulfides (total captopril). For total captopril analysis, disulfides were reduced with tributylphosphine (TBP) prior to protein precipitation. Since the reducing agent interfered with the MBB derivatization reaction, this method was not suitable for total captopril analysis. Both methods were validated for the bioanalysis of free captopril in human plasma. After removal of plasma proteins, samples were analyzed without an additional extraction procedure. The limit of quantitation in plasma was 12.5 ng/ml for the MBB method (limit of detection 30 pg) and 25 ng/ml for the OPA method (limit of detection 50 pg). The OPA method was also validated for total captopril analysis in human plasma and urine. The limit of quantitation was 25 ng/ml in plasma and 250 ng/ml in urine (limit of detection 50 pg). We conclude that for the analysis of free captopril the precolumn MBB method is superior to the OPA method since only the derivatization reaction has to be carried out immediately. The postcolumn OPA method is especially suitable for the analysis of total captopril since reducing reagents and high concentrations of endogenous thiols do not interfere with the derivatization reaction.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/blood , Antihypertensive Agents/blood , Captopril/blood , Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Bridged Bicyclo Compounds , Captopril/pharmacokinetics , Chromatography, High Pressure Liquid , Fluorescent Dyes , Humans , Male , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Fluorescence , o-Phthalaldehyde
13.
Kidney Int ; 52(6): 1693-9, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9407519

ABSTRACT

A renal-specific controlled release of an active drug may enable a reduction of the required dose and may provide a reduction of extra-renal toxicity. To achieve renal specific targeting of the NSAID naproxen, the low-molecular-weight protein (LMWP) lysozyme was employed as carrier since it is mainly taken up and catabolized in the proximal tubules of the kidney. A conjugate was synthesized with an average coupling degree of 2 mol naproxen per 1 mol lysozyme in which the drug was directly coupled to the protein via a peptide bond. First, we investigated whether naproxen conjugation affects the renal disposition of lysozyme. As native lysozyme, the conjugate was predominantly and rapidly (within 20 min) taken up by the kidney. The subsequent decrease in renal content reflecting the renal degradation of the conjugated lysozyme molecules appeared also to be similar to that of native lysozyme with a half life of four hours. Second, the effect of lysozyme conjugation on the body distribution of naproxen was studied. An important observation with regard to the aimed reduction in extra-renal side effects was that no detectable amounts of free naproxen were present in the plasma after administration of conjugate. Conjugation of naproxen to lysozyme resulted in a pronounced (70-fold) increase of naproxen accumulation in the kidney. In agreement with the protein disposition study, the conjugate was rapidly taken up by the kidney and subsequently degraded. In conclusion, renal selective targeting of the NSAID naproxen can be obtained by conjugation with the LMWP lysozyme. This concept of drug delivery to the kidney has the potential to improve drug efficacy and safety.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Kidney Diseases/drug therapy , Muramidase/pharmacokinetics , Naproxen/pharmacokinetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Dinoprostone/urine , Female , Iodine Radioisotopes , Kidney Tubules, Proximal/metabolism , Kinetics , Male , Muramidase/chemical synthesis , Naproxen/chemical synthesis , Rats , Rats, Wistar , Tissue Distribution
14.
Pharm Res ; 10(7): 963-9, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8378258

ABSTRACT

Low molecular weight proteins (LMWPs) are potential carriers for targeting drugs to the kidney. To test whether ester bonds are suitable for the reversible drug conjugation, the antiinflammatory drug naproxen (Nap) was conjugated to the LMWP lysozyme (LYSO) via an ester bond using an L-lactic acid spacer (Nap-lact-LYSO, 1:1:1). The distribution and degradation of the conjugate in rats were compared to those of an equimolar mixture of free drug and LMWP and of a directly coupled conjugate without spacer (Nap-LYSO). The plasma clearance of Nap-lact-LYSO closely resembled that of Nap-LYSO and LYSO itself. Its major accumulation site appeared to be the kidney as demonstrated by extracorporal gamma-camera counting of the LMWP. Renally released naproxen was excreted in the urine as 6-desmethyl-naproxen-sulfate (6-DMN-S). Apparently the kidneys represent the main sites of demethylation and sulfation after administration of the LMWP-coupled drug. In addition, the renal excretion of naproxen (including its metabolites) was significantly delayed and sustained as compared to that after injection of uncoupled naproxen. Using the L-lactic acid spacer LMWP conjugation, the renal selectivity of Nap was increased 5.6 +/- 0.41-fold. Additional in vitro studies with Nap-lact-LYSO revealed that renal generation of the parent drug coincided with formation of low molecular weight catabolites, mainly as naproxen-L-lactic acid-lysine (Nap-lact-Lys). This indicated that in vitro the rate of cleavage of the ester bond is significantly slower than digestion of the carrier backbone itself.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Kidney/metabolism , Lactates/chemistry , Muramidase/chemistry , Naproxen/administration & dosage , Anesthesia , Animals , Drug Carriers , Gamma Cameras , Iodine Radioisotopes , Isotope Labeling , Kidney Cortex/metabolism , Lactic Acid , Lysosomes/metabolism , Male , Molecular Weight , Naproxen/chemistry , Naproxen/pharmacokinetics , Rats , Rats, Wistar , Tissue Distribution
16.
Pharm Weekbl Sci ; 14(4): 191-5, 1992 Aug 21.
Article in English | MEDLINE | ID: mdl-1437498

ABSTRACT

In this report we describe the conditions of collection, storage and handling of urine samples, collected after oral dosing with indomethacin in man, in order to maintain the integrity of the labile glucuronide formed. We found that the body clearance occurs predominantly by renal metabolism, due to glucuronidation in the human kidney. These glucuronides may be converted to isomeric glucuronides and/or the parent compound indomethacin during the residence time in the bladder.


Subject(s)
Glucuronates/metabolism , Indomethacin/pharmacokinetics , Kidney/metabolism , Adult , Chromatography, High Pressure Liquid , Drug Stability , Glucuronates/pharmacokinetics , Humans , Hydrogen-Ion Concentration , Indomethacin/urine , Male , Temperature
17.
J Med Chem ; 35(7): 1246-59, 1992 Apr 03.
Article in English | MEDLINE | ID: mdl-1560438

ABSTRACT

Low molecular weight proteins (LMWPs) are known to be reabsorbed and catabolized primarily by the proximal tubular cells of the kidneys. As such, LMWPs might serve as drug carriers that release drugs site-specifically in the kidney. We tested this concept in vitro by coupling different drugs to the LMWP lysozyme both directly (amide bond) and via different spacers: oligopeptides (amide bond), (poly-)alpha-hydroxy acids (ester bond), and a pH sensitive cis-aconityl spacer (amide bond). The capability of the kidney to release the parent drug from such drug-spacer derivatives and drug-LMWP conjugates by enzymatic or chemical hydrolysis of the bond was tested by incubation experiments in renal cortex homogenates and lysosomal lysates. Directly coupled conjugates of terminal carboxyl group containing drugs and lysozyme were catabolized to single amino acids, but did not result in release of the parent drug. The amide bond between the drug and the final amino acid (lysine) appeared to be stable in the incubation milieu. Different oligopeptide spacers coupled to the drugs showed similar results: the oligopeptide itself was cleaved but the amide bond between the drug and different single amino acids remained untouched. Only amide bonds of derivatives of carboxylic drugs with peptide structures themselves were cleaved. Some of the directly coupled conjugates of terminal amino drugs and oligopeptides showed clear release of the parent drug whereas others were stable. Terminal amino drugs were rapidly released from an acid-sensitive cis-aconityl spacer. Terminal carboxyl group containing drugs were enzymatically released from their glycolic and lactic ester spacers at different rates. These kinds of drugs were also released as parent drug from LMWP conjugates with ester spacers like L-lactic acid. Increasing spacer length by intercalating a tetra(L-lactic acid) molecular between the drug and the protein further increased the extent and rate of drug release, indicating increased accessibility of the bond to the enzymes. Terminal amino group containing drugs were rapidly generated as parent drug from LMWP conjugates using an acid-sensitive spacer. In addition the conjugates were found to be adequately stable in plasma, considering their rapid clearance from the bloodstream. It is concluded that LMWPs may indeed be of use as carriers for specific renal delivery of drugs, since renal cortex homogenates and lysosomal lysates are able to catabolize the protein and generate the parent drug from drug-LMWP conjugates bearing suitable spacers. The option of enzymatic release is limited by the narrow specificity of the lysosomal enzymes.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Drug Carriers/chemistry , Kidney Cortex/metabolism , Lysosomes/metabolism , Proteins/chemistry , Amino Acids/metabolism , Animals , Chromatography, High Pressure Liquid , Endopeptidases/metabolism , Hydrogen-Ion Concentration , Lactates/chemistry , Lactic Acid , Leucine/chemistry , Male , Molecular Weight , Muramidase/chemistry , Muramidase/metabolism , Naproxen/administration & dosage , Naproxen/pharmacokinetics , Oligopeptides/chemistry , Oligopeptides/metabolism , Proteins/metabolism , Rats , Rats, Inbred Strains , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Succinimides
18.
Pharm Res ; 8(10): 1223-30, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1796038

ABSTRACT

Low molecular weight proteins (LMWPs), such as lysozyme, may be suitable carriers to target drugs to the kidney. In this study the antiinflammatory drug naproxen was covalently bound to lysozyme (1:1). Pharmacokinetics of the conjugate, naproxen-lysozyme (nap-LYSO), were compared to that of an equimolar mixture of uncoupled naproxen with lysozyme in freely moving rats. Similar plasma kinetics and organ distribution for native lysozyme and the drug conjugate were observed (Clp = 1.2 and 1.1 ml/min; t1/2,beta = 85 and 75 min, respectively). In case of the uncoupled naproxen-lysozyme mixture, a monoexponential plasma disappearance of naproxen with a t1/2 of 2.8 hr was observed, coinciding with urinary excretion of naproxen metabolites (mainly 6-desmethylnaproxen sulfate; 6-DMN-S) between 2 and 8 hr after injection. Urinary recovery of total metabolites was 59% of the naproxen dose. In contrast, after injection of covalently bound naproxen, plasma levels of the parent drug were below the detection level, whereas naproxen was recovered as 6-DMN-S in urine over a period from 4 to 30 hr. However, only 8% of the administered dose was recovered as 6-DMN-S in urine, whereas 50% of the dose was recovered as naproxen metabolites in feces. Incubation experiments using purified renal tubular lysosomal lysates revealed that naproxen-lysozyme degradation ultimately results in a stable naproxen amino acid catabolite, naproxen-lysine (nap-lys). Hepatic uptake and biliary excretion of this catabolyte were demonstrated in isolated perfused rat livers. Further, an equipotent pharmacological activity relative to parent naproxen was observed.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Carrier Proteins/chemistry , Kidney/drug effects , Muramidase/chemistry , Naproxen/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cyclooxygenase Inhibitors/pharmacology , Drug Carriers , Guinea Pigs , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Lysine/chemistry , Male , Molecular Weight , Muramidase/administration & dosage , Naproxen/administration & dosage , Naproxen/analogs & derivatives , Naproxen/pharmacokinetics , Protein Binding , Rats , Rats, Inbred Strains , Tissue Distribution
19.
J Med Chem ; 34(3): 1137-41, 1991 Mar.
Article in English | MEDLINE | ID: mdl-2002455

ABSTRACT

In order to investigate whether neoglycoproteins can potentially act as carriers for targeting of antiviral drugs to certain cell types in the body, various neoglycoproteins were synthesized using thiophosgene-activated p-aminophenyl sugar derivatives. These neoglycoproteins were conjugated with the 5'-monophosphate form of the antiviral drug AZT. For a proper characterization of these preparations, both protein and drug content have to be determined. Comparison of the Lowry and the Bio-Rad protein assays revealed that for both the neoglycoprotein carriers themselves and the AZTMP conjugates, the Lowry assay yielded the most reliable and reproducible results. It was demonstrated that both the reagent used for drug conjugation (ECDI) as well as the introduction of phenyl-sugar groups in the protein interfered with the analysis of bound nucleotide as based on spectral differences between protein and protein-drug conjugate. Therefore, we developed a rapid HPLC system for determination of the drug-protein coupling ratio through acid hydrolysis of the covalently bound nucleotide. With the ECDI-mediated conjugation of 5'-monophosphate drug derivatives to neoglycoproteins, products with molar ratios of drug to protein ranging from 1.2 to 5.6 were obtained. The drug-neoglycoprotein conjugates appeared to be fairly stable during storage, in lyophilized form, at -20 degrees C. The anti-HIV-1 activity of the neoglycoprotein-drug conjugates, as determined in vitro in MT-4 cells, was shown to be dependent on glycosylation of the albumin and also on the kind of sugar present in the neoglycoprotein. The anti-HIV-1 activity of the AZTMP-mannose-albumin conjugate exceeded that of the parent drug by more than 4 times.


Subject(s)
Antiviral Agents/chemistry , Glycoproteins/chemical synthesis , Thymine Nucleotides/chemistry , Zidovudine/analogs & derivatives , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Dideoxynucleotides , Drug Carriers , Drug Stability , Glycoproteins/analysis , Glycoproteins/pharmacology , HIV-1/drug effects , Hydrolysis , Mannose/chemistry , Mannose/metabolism , Mannose/pharmacology , Proteins/analysis , Serum Albumin/chemistry , Serum Albumin/metabolism , Serum Albumin/pharmacology , Thymine Nucleotides/administration & dosage , Thymine Nucleotides/pharmacology , Zidovudine/administration & dosage , Zidovudine/chemistry , Zidovudine/metabolism , Zidovudine/pharmacology
20.
J Biol Chem ; 266(5): 3343-8, 1991 Feb 15.
Article in English | MEDLINE | ID: mdl-1993707

ABSTRACT

We synthesized several para-aminophenyl (pap-) mannose-terminated albumins with varying sugar density (Man7-HSA, Man22-HSA, and Man40-HSA) and compared hepatic uptake with (thio-)mannose-terminated bovine serum albumin (Man-43-AI-BSA) The rate of uptake in isolated perfused rat livers was found to be positively correlated with the sugar density (Man40-HSA = Man22-HSA greater than Man7-HSA greater than HSA). Immunohistochemical staining of liver sections showed for both types of neoglycoproteins that uptake occurred in nonparenchymal cells only. Competition experiments with a 500-fold excess of mannan, a known ligand for the mannose/N-acetylglucosamine receptor, that is predominantly localized in endothelial cells, showed complete inhibition of the (thio-)Man43-AI-BSA uptake. In the case of (pap-)mannose-terminated albumins, however, the extent of inhibition by mannan was moderate and decreased markedly with increasing sugar density, being only 20% for (pap-)Man40-HSA. Therefore, we hypothesized that one or more additional removal systems contributed to the clearance of these (pap-)mannose glycoproteins. We found that net negative charge of the (pap-)mannose albumins clearly increased with increasing sugar density, as shown on fast protein liquid chromatography anion-exchange chromatograms. To determine whether the scavenger receptor system that is also mainly present on endothelial cells is involved, we performed competition studies with strongly negatively charged substrates, such as dextran sulfate and formaldehyde-treated human serum albumin (fHSA). An excess of dextran sulfate (500 kDa), indeed blocked the (pap-)mannose-albumin uptake for more than 95%. Dextran sulfate completely inhibited the hepatic uptake of mannan as well, indicating that the polyanion does not discriminate between the scavenger system and the mannose receptor system and should be regarded as an aspecific inhibitor of receptor-mediated endocytotic pathways. Surprisingly, a 500-fold excess of fHSA only moderately (20%) inhibited the clearance of (pap-)Man40-HSA in spite of its high affinity for the scavenger receptor. However, a combination of mannan and fHSA strongly inhibited the uptake of (pap-)Man22-HSA (90%) and to a lesser extent (pap-)Man40-HSA (80%), indicating that a third uptake mechanism may exist that recognizes both mannose groups (or other sugars) and net negative charge. This so far unnoticed receptor system apparently is strongly affected by dextran sulfate and, as shown by immunohistochemistry, is mainly localized on Kupffer cells rather than on the endothelial cells of the liver.


Subject(s)
Albumins/metabolism , Carbohydrate Metabolism , Endocytosis , Lectins, C-Type , Liver/cytology , Mannose-Binding Lectins , Mannose/metabolism , Receptors, Cell Surface , Animals , Chromatography, Liquid , Dextran Sulfate/metabolism , Endothelium/metabolism , Immunohistochemistry , In Vitro Techniques , Kupffer Cells/metabolism , Liver/metabolism , Male , Mannose Receptor , Rats , Rats, Inbred Strains , Receptors, Immunologic/metabolism , Serum Albumin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...