Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Pharm Res ; 37(7): 138, 2020 Jul 10.
Article in English | MEDLINE | ID: mdl-32651732

ABSTRACT

PURPOSE: Forced degradation is critical to probe the stabilities and chemical reactivities of therapeutic peptides. Typically performed in bulk followed by LC-UV or LC-MS analysis, this traditional workflow consists of a reaction/analysis sequence and usually requires half a day to several days to form and measure the desired amounts of degradants. A faster method is needed to study peptide degradation in a shorter time in order to speed up the drug development process. METHODS: In the new rapid method developed in this study, peptide degradation occurs in levitated aqueous microdroplets using the Leidenfrost effect. RESULTS: This two-minute reaction/analysis workflow allows major degradation pathways of Buserelin, Octreotide, Desmopressin and Leuprorelin to be studied. The reactions include deamidation, disulfide bond cleavage, ether cleavage, peptide bond hydrolysis, and oxidation. CONCLUSIONS: The accelerated forced degradation method requires a minimal amount of therapeutic peptide per stress condition, and the appropriate extent of degradation can be readily generated in seconds by adjusting the droplet levitation time. Levitated microdroplets should be applicable in pharmaceutical development to rapidly determine the intrinsic stability of therapeutic peptides and to aid formulation development by screening the effects of excipients on the stability of the peptides. Graphical abstract.


Subject(s)
Buserelin/chemistry , Deamino Arginine Vasopressin/chemistry , Leuprolide/chemistry , Octreotide/chemistry , Drug Compounding , Drug Stability , Hot Temperature , Hydrogen-Ion Concentration , Kinetics , Particle Size , Protein Stability , Proteolysis , Workflow
2.
Protein Pept Lett ; 26(9): 691-701, 2019.
Article in English | MEDLINE | ID: mdl-31215364

ABSTRACT

BACKGROUND: Both biodegradable and non-biodegradable peptide-loaded implants are already developed for the long-term treatment of patients, thereby reducing the frequency of drug administration. To further improve peptide formulation, extending the scope of implant-based drug delivery systems towards other polymers and processing techniques is highly interesting. OBJECTIVE: In this study, as a proof-of-principle, the feasibility of hot-melt processing of a peptide active pharmaceutical ingredient was assessed by developing a non-biodegradable poly(ethylenevinyl acetate) (33% VA) implant loaded with 20% (w/w) buserelin acetate. METHODS: Cross-sectional implant characterization was performed by Raman microscopy. The stability of buserelin acetate in the polymeric matrix was evaluated for 3 months under ICH stability conditions and the quantity as well as the degradation products analyzed using LC-UV methods. An in vitro dissolution study was performed as well and buserelin acetate and its degradants analyzed using the same chromatographic methods. RESULTS: No significant quantities of buserelin acetate-related degradation products were formed during the hot-melt preparation as well as during the stability study. Together with the consistent buserelin acetate assay values over time, chemical peptide stability was thus demonstrated. The in vitro buserelin acetate release from the implant was found to be diffusion-controlled after an initial burst release, with stable release profiles in the stability study, demonstrating the functional stability of the peptide implant. CONCLUSION: These results indicate the feasibility of preparing non-biodegradable peptide-loaded implants using the hot-melt production method and may act as a proof of principle concept for further innovation in peptide medicinal formulations.


Subject(s)
Buserelin/chemistry , Drug Carriers/chemistry , Drug Implants/chemistry , Polyvinyls/chemistry , Buserelin/administration & dosage , Drug Compounding/methods , Drug Implants/administration & dosage , Drug Liberation , Drug Stability , Feasibility Studies , Hot Temperature , Proof of Concept Study , Solubility
3.
J Liposome Res ; 26(1): 69-79, 2016.
Article in English | MEDLINE | ID: mdl-25945393

ABSTRACT

CONTEXT: Oral delivery of peptide and protein drugs still remains the area of challenges due to their low stability and permeability across GI tract. Among numerous attempts, the receptor-mediated drug targeting is a promising approach to enhance GI permeability. OBJECTIVE: The aim of this study was to prepare mannosylated buserelin acetate (MANS-BA) proliposome powders grafted with N-octadecyl-d-mannopyranosylamine (SAMAN) as targeting moiety and evaluate their permeability across Caco-2 cell monolayers. MATERIALS AND METHODS: The MANS-BA proliposome powders were prepared by coprecipitation method. The targeting moiety SAMAN was synthesized in-house and confirmed by characterization using Fourier transform infrared (FTIR) and differential scanning calorimeter (DSC). RESULTS: The MANS-BA liposomes reconstituted from proliposome powders exhibited the oligolamellar vesicular structure of phospholipid bilayer. Their size, zeta potential and entrapment efficiency were in the ranges of 93.11-218.95 nm, -24.03 to -37.15 mV and 21.12-33.80%, respectively. The permeability of reconstituted MANS-BA liposomes across Caco-2 cell monolayers was significantly enhanced to about 1.2- and 2.2-fold over those of conventional BA liposomes and solution, respectively. DISCUSSION: Increase in dicetylphosphate, cholesterol and SAMAN contents resulted in significant increase in size and zeta potential of reconstituted MAN-BA liposomes. The entrapment efficiency was increased with increasing dicetylphosphate and mannitol contents in liposomes containing cholesterol. CONCLUSIONS: The significantly enhanced permeability across Caco-2 cell monolayers of MANS-BA liposomes might be due to the role of mannose receptor on intestinal enterocytes.


Subject(s)
Amino Sugars/chemistry , Buserelin/chemistry , Liposomes/chemistry , Amino Sugars/chemical synthesis , Buserelin/chemical synthesis , Caco-2 Cells , Humans , Ligands , Liposomes/chemical synthesis , Permeability
4.
Int J Pharm ; 467(1-2): 48-9, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24657556

ABSTRACT

Buserelin is a GnRH agonist peptide drug, comprising a nine amino acid sequence (pGlu-His-Trp-Ser-Tyr-D-Ser(tBu)-Leu-Arg-Pro-NH-Et) and most commonly known for its application in hormone dependent cancer therapy, e.g. prostate cancer. In order to evaluate its hot-melt extrusion (HME) capabilities, buserelin powder in its solid state was exposed to elevated temperatures for prolonged time periods. A stability indicating UPLC-PDA method was used for quantification of buserelin and the formed degradants. Different solid state kinetic models were statistically evaluated of which the Ginstling-Brounshtein model fitted the data best. Extrapolation to and experimental verification of typical HME-related conditions, i.e. 5 min at 100°C and 125°C, showed no significant degradation, thus demonstrating the HME capabilities of buserelin. Mass spectrometric identification of the buserelin-related degradants formed under solid state heat stress was performed. Based upon the identity of these degradants, different degradation hypotheses were raised. First, direct ß-elimination of the hydroxyl moiety at the serine residue, followed by fragmentation into an amide (pGlu-His-Trp-NH2) and pyruvoyl (pyruvoyl-Tyr-D-Ser(tBu)-Leu-Arg-Pro-NH-Et) peptide fragments, was postulated. Alternatively, internal esterification due to nucleophilic attack of the unprotected serine residue, followed by ß-elimination or hydrolysis would yield pGlu-His-Trp, pGlu-His-Trp-NH2 and the pyruvoyl peptide fragment. Degradant pGlu-His-Trp-Ser-Tyr-NH2 is believed to be formed in a similar way. Secondly, direct backbone hydrolysis would yield pGlu-His-Trp and Tyr-D-Ser(tBu)-Leu-Arg-Pro-NH-Et peptide fragments. Moreover, the presence of Ala-Tyr-D-Ser(tBu)-Leu-Arg-Pro-NH-Et can be explained by hydrolysis of the Trp-Ser peptide bond and conversion of the serine moiety to an alanine moiety. Third and finally, isomerisation of aforementioned peptide fragments and buserelin itself was also observed.


Subject(s)
Antineoplastic Agents, Hormonal/chemistry , Buserelin/chemistry , Hot Temperature , Chromatography, High Pressure Liquid , Drug Stability , Hydrolysis , Isomerism , Kinetics , Models, Chemical , Molecular Structure , Powders , Protein Stability , Proteolysis , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Technology, Pharmaceutical/methods
5.
Int J Pharm ; 339(1-2): 130-8, 2007 Jul 18.
Article in English | MEDLINE | ID: mdl-17398044

ABSTRACT

We investigated the effect of different dispersion methods on release behavior and efficacy onset following microparticle administration of buserelin acetate (BA) sustained-release injection. In this in vitro release study, the initial dispersion of BA increased with increased stirring speed (p<0.01). Stability of BA was studied over 7 days after BA release. The initial BA release rate was higher (p<0.01) after a 1-min vibration dispersion method (VDM) using a test tube mixer (2000 rpm) compared with the standard dispersion method (SDM) by hand. Without shaking, powder aggregation was observed, and BA release was lower than in either the SDM or VDM methods. In this study using 4-week-old Sprague-Dawley female rats, the initial plasma estrone (E(1)) concentrations were lower (p<0.05) in the VDM method than in the SDM method. Observations by optical microscope and scanning microscope showed no change in microparticle shape or distribution of size induced by SDM, VDM or the ultrasonication dispersion method. These results suggest that different dispersion methods do not change the shape and distribution of microparticle size, but clearly change the BA release rate and the transition in plasma E(1) concentrations that can affect drug efficacy.


Subject(s)
Buserelin/administration & dosage , Estrone/blood , Animals , Buserelin/analysis , Buserelin/chemistry , Chromatography, High Pressure Liquid , Down-Regulation , Female , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Microscopy, Electron, Scanning , Microspheres , Rats , Rats, Sprague-Dawley , Solubility
6.
Int J Pharm ; 332(1-2): 107-14, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17084049

ABSTRACT

The objective of this study was to investigate the in vitro drug (diltiazem hydrochloride and buserelin acetate) release from different in situ forming biodegradable drug delivery systems, namely polymer solutions (in situ implants) and in situ microparticle (ISM) systems. The drug release from ISM systems [poly(d,l-lactide) (PLA) or poly(d,l-lactide-co-glycolide) (PLGA)-solution dispersed into an external oil phase] was investigated as a function of the type of solvent and polymer, polymer concentration and internal polymer phase:external oil phase ratio and was compared to the drug release from in situ implant systems and microparticles prepared by conventional methods (solvent evaporation or film grinding). Upon contact with the release medium, the internal polymer phase of the ISM system solidified and formed microparticles. The initial drug release from ISM systems decreased with increasing polymer concentration and decreasing polymer phase:external oil phase ratio. The type of biocompatible solvent also affected the drug release. It decreased in the rank order DMSO>NMP>2-pyrrolidone. In contrast to the release of the low molecular weight diltiazem hydrochloride, the peptide release (buserelin acetate) was strongly dependent on the polymer degradation/erosion. One advantage of the ISM system when compared to in situ implant systems was the significantly reduced burst effect because of the presence of an external oil phase. ISM systems resulted in drug release profiles comparable to the drug release of microparticles prepared by the solvent evaporation method. Therefore, the ISM systems are an attractive alternative to existing complicated microencapsulation methods.


Subject(s)
Buserelin , Diltiazem , Drug Carriers , Drug Delivery Systems , Lactic Acid/chemistry , Polyglactin 910/chemistry , Polymers/chemistry , Solvents/chemistry , Buserelin/administration & dosage , Buserelin/chemistry , Chemistry, Pharmaceutical , Delayed-Action Preparations , Diltiazem/administration & dosage , Diltiazem/chemistry , Dimethyl Sulfoxide/chemistry , Drug Compounding , Drug Implants , Emulsions , Kinetics , Models, Chemical , Molecular Weight , Particle Size , Pharmaceutical Solutions , Polyesters , Porosity , Pyrrolidinones/chemistry , Solubility , Technology, Pharmaceutical
8.
Pharm Res ; 15(9): 1449-55, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9755899

ABSTRACT

PURPOSE: To develop a method for calculating epimerisation parameters, find out if the kinetics of the independent reactions can be established, and elucidate primary structure-chemical degradation relationships in the degradation kinetics of three gonadorelin analogues. METHODS: The influences of pH, temperature, and buffer concentration on the degradation of the three gonadorelin analogues buserelin, goserelin, and triptorelin were investigated using RP-HPLC. A method was developed to calculate epimerisation and hydrolysis rate constants independently. RESULTS: Explicit structure-degradation mechanism relations were found in the degradation of all three compounds. The L-serine residue was found to be involved in both a solvent-catalysed backbone hydrolysis and a hydroxyl-catalysed epimerisation whereas, the O-tertiary butyl D-serine residue was only involved in proton-catalysed ether hydrolysis. The kinetics of identical reactions in different analogues were generally comparable. CONCLUSIONS: The degradation of the gonadorelin analogues is located at a relatively small number of chemical residues and prediction of the degradation mechanisms and kinetics of other peptides with similar structural elements appears to be possible.


Subject(s)
Buserelin/chemistry , Gonadotropin-Releasing Hormone/analogs & derivatives , Goserelin/chemistry , Triptorelin Pamoate/chemistry , Buffers , Chromatography, High Pressure Liquid , Hydrogen-Ion Concentration , Kinetics , Models, Chemical , Protein Conformation , Temperature
9.
Pharm Res ; 14(10): 1401-5, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9358553

ABSTRACT

PURPOSE: The present study addresses how maltosyl-beta-cyclodextrin (G2-beta-CyD) impacts upon the alpha-chymotrypsin-catalyzed hydrolysis of buserelin acetate, an agonist of luteinizing hormone-releasing hormone with emphasis upon the direct effect of G2-beta-CyD on the activity of the protease. METHODS: Kinetic and solubility studies were performed in isotonic phosphate buffer (pH 7.4) at 25 degrees C and 37 degrees C. The interaction of alpha-chymotrypsin with G2-beta-CyD in the buffer solution was examined by differential scanning calorimetry. RESULTS: G2-beta-CyD decelerated the alpha-chymotrypsin-catalyzed hydrolysis of buserelin acetate to give the 1-3 tripeptide and the 4-9 hexapeptide fragments. This deceleration can be explained solely by a non-productive encounter between a complex of the substrate with G2-beta-CyD and the protease at relatively low CyD concentrations, while the direct inhibitory effect of G2-beta-CyD on the proteolytic activity made a considerable contribution to the overall deceleration of the hydrolysis at higher CyD concentrations. Calorimetric studies indicate the presence of intermediate states in the thermal unfolding of alpha-chymotrypsin, simultaneously accompanied by the autolysis. By contrast, a two-state thermal unfolding of alpha-chymotrypsin was observed in the presence of G2-beta-CyD, suggesting reduced proteolytic activity upon binding to G2-beta-CyD. CONCLUSIONS: These results suggest that G2-beta-CyD at higher concentrations inhibits the proteolytic action of alpha-chymotrypsin through direct interaction with the protease, as well as through the formation of a non-productive complex with the substrate.


Subject(s)
Buserelin/metabolism , Chymotrypsin/antagonists & inhibitors , Cyclodextrins/pharmacology , Enzyme Inhibitors/metabolism , Gonadotropin-Releasing Hormone/agonists , beta-Cyclodextrins , Buserelin/chemistry , Calorimetry, Differential Scanning , Catalysis , Chromatography, High Pressure Liquid , Chymotrypsin/chemistry , Cyclodextrins/chemistry , Enzyme Inhibitors/chemistry , Hydrolysis , Kinetics , Protein Denaturation
10.
FEBS Lett ; 391(1-2): 162-6, 1996 Aug 05.
Article in English | MEDLINE | ID: mdl-8706907

ABSTRACT

The sexual hormone of S. cerevisiae, alpha-mating factor (alpha-MF, WHWLQLKPGQPMY) has structural homology with mammalian luteinizing hormone releasing hormone (LHRH, pEHWSYGLRPG-NH2) and has been shown to exhibit LHRH activity [Loumaye et al. (1982) Science 218, 1323-1325]. We have tested whether LHRH has alpha-MF activity in yeast and found that it does not. We therefore synthesized a series of hybrid peptides of alpha-MF and LHRH to study the structural features which determine alpha-MF and LHRH activities. A hybrid peptide consisting of the LHRH sequence with the C-terminal tetrapeptide (QPMY) of alpha-MF did not exhibit alpha-MF activity. Thus, the lack of alpha-MF activity of LHRH is not due solely to the absence of the C-terminal residues. Substitution of Lys7 in alpha-MF with Arg, as is found in LHRH, did not affect the alpha-MF activity, nor did an additional substitution of Trp1 with pGlu. However, the C-terminal four amino acids of alpha-MF were necessary for alpha-MF activity. Our results indicate that insertion of a Ser residue in position 4 as found in LHRH abolishes alpha-MF activity. These results suggest that, in addition to an intact C-terminus, correct spacing of the N-terminal His2 and the C-terminus is required for alpha-MF activity. The hybrid peptides all exhibited less LHRH activity than either LHRH or alpha-MF. These structure-function studies indicate that the structural homology between these two reproductive hormones may not reflect an evolutionary relationship between them.


Subject(s)
Peptides/chemistry , Peptides/metabolism , Receptors, LHRH/metabolism , Saccharomyces cerevisiae/physiology , Amino Acid Sequence , Animals , Buserelin/chemistry , Cell Membrane/metabolism , Chickens , Fishes , Gonadotropin-Releasing Hormone/chemistry , Mammals , Mass Spectrometry , Mating Factor , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/pharmacology , Pheromones/chemistry , Pheromones/metabolism , Saccharomyces cerevisiae/drug effects , Sequence Homology, Amino Acid
11.
Int J Pept Protein Res ; 48(1): 59-70, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8844264

ABSTRACT

The S- and C-glycosylated nonapeptides 1 and 2 were synthesized as analogs of the non-glycosylated LH-RH agonist buserelin (pGlu-His-Trp-Ser-Tyr-D-Ser(tBu)-Leu-Arg-Pro-NHEt) by segment condensation in solution. 1 and 2 differ from this peptide in the amino acid in position 6. In the first case (1), D-serine (tBu) is substituted by D-cysteine carrying a rhamnosyl residue, in the second case (2) D-alanine carrying a galactosyl moiety bound as C-glycoside is incorporated. The bioactivity of both glycopeptides as fertility drugs was determined from the dose dependent LH release in male rats. Additionally, in female rats the ovulation rate was assessed. As a result the analog 1 exhibits a similar biological activity as buserelin while analog 2 shows about 25% of this potency. Compared to buserelin the solubility of the analogs 1 and 2 in aqueous buffer is improved by more than two orders of magnitude due to the carbohydrate moieties.


Subject(s)
Buserelin/analogs & derivatives , Buserelin/chemistry , Glycopeptides/chemical synthesis , Gonadotropin-Releasing Hormone/agonists , Alanine/chemistry , Animals , Buserelin/pharmacology , Carbohydrates/chemistry , Cysteine/chemistry , Female , Luteinizing Hormone/blood , Luteinizing Hormone/drug effects , Luteinizing Hormone/metabolism , Male , Ovulation/drug effects , Protein Conformation , Rats , Structure-Activity Relationship
12.
Gynecol Endocrinol ; 8(3): 215-22, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7847108

ABSTRACT

This review addresses the question of whether the different gonadotropin releasing hormone (GnRH) agonists in clinical use might have different impacts, related to their chemical structure, delivery system and dose. Impact was investigated in benign gynecological disorders, i.e. endometriosis and leiomyoma. Arguments are presented indicating that a difference in impact of different analogs can be expected. All currently used intranasal, daily subcutaneous and depot preparations finally give rise to low levels of serum estradiol. The number of days before the first ovulatory menstruation after discontinuation of GnRH agonist treatment is remarkably constant. Four weeks after the last impact of the agonist, there is resumption of follicle growth. This phenomenon is independent of chemical structure, delivery system and dose. One should realize, however, that it generally takes about 30 days before the impact of a depot preparation disappears. Consequently, the impact of a depot preparation lasts 4 weeks longer than that of an otherwise applied agonist. Thus resumption of pituitary activity after discontinuation of a depot formulation takes 4 weeks longer than after discontinuation of non-depot formulations. All agonists have an impressive effect on endometriosis, independent of their chemical structure and delivery system. However, there are no studies comparing different agonists with the same delivery system in comparable endometriosis groups. Similarly, all agonists considerably reduce myoma volume, independently of their chemical structure and delivery system.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Endometriosis/drug therapy , Gonadotropin-Releasing Hormone/agonists , Leiomyoma/drug therapy , Buserelin/chemistry , Buserelin/pharmacokinetics , Buserelin/therapeutic use , Dose-Response Relationship, Drug , Drug Delivery Systems , Estradiol/blood , Female , Gonadotropin-Releasing Hormone/pharmacokinetics , Goserelin/chemistry , Goserelin/pharmacokinetics , Goserelin/therapeutic use , Humans , Leuprolide/chemistry , Leuprolide/pharmacokinetics , Leuprolide/therapeutic use , Menstrual Cycle/drug effects , Nafarelin/chemistry , Nafarelin/pharmacokinetics , Nafarelin/therapeutic use , Structure-Activity Relationship , Triptorelin Pamoate/chemistry , Triptorelin Pamoate/pharmacokinetics , Triptorelin Pamoate/therapeutic use
13.
Dtsch Med Wochenschr ; 119(14): 523-4, 1994 Apr 08.
Article in German | MEDLINE | ID: mdl-8156885
14.
Boll Chim Farm ; 133(3): 156-9, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8011275

ABSTRACT

Obtaining and some properties of suspensions with buserelin-there were obtained buserelin suspensions with zinc ions with pH 7.2-7.5 and hormone: zinc molar ratio from 1:2.5 to 1:500. The obtained buserelin suspensions show moderated hormone release in vitro and prolonged action in vivo. The pulsating buserelin release from suspensions has been found in vitro and in vivo.


Subject(s)
Buserelin/administration & dosage , Animals , Buserelin/chemistry , Buserelin/pharmacokinetics , Chemical Phenomena , Chemistry, Physical , Injections, Intramuscular , Suspensions , Swine
15.
Ann N Y Acad Sci ; 672: 510-27, 1992 Nov 30.
Article in English | MEDLINE | ID: mdl-1476387

ABSTRACT

Amino acids play an important role in biochemistry and chemistry. They are the building blocks of proteins and play an essential role in the regulation of the metabolism of living organisms. In general, it can be stated that microbial processes (fermentation) are the industrial production methods of choice for large-scale production of naturally occurring proteinogenic L-alpha-H-amino acids, while for the production of synthetic D- and/or L-alpha-H-amino acids, several other methods are highly competitive. At DSM, several routes, i.e., (chemoenzymatic) synthesis, towards L-alpha-H and D-alpha-H-amino acids have been elaborated since the midseventies. A general process for the synthesis of natural as well as synthetic optically pure amino acids has been developed, using an enzymatic kinetic resolution step on racemic amino acid amides as the key step. In this case, both enantiomers of the alpha-H-amino acids are prepared in one single step. This process has been commercialized since 1988. More recent developments using L- or D-amino peptidases in combination with amino acid amide racemases and an asymmetric transformation concept are discussed.


Subject(s)
Amino Acids/chemical synthesis , Peptides/chemical synthesis , Amino Acid Sequence , Amino Acids/chemistry , Buserelin/chemistry , Gonadotropin-Releasing Hormone/chemistry , Kinetics , Leucyl Aminopeptidase/metabolism , Leuprolide/chemistry , Molecular Sequence Data , Pseudomonas putida/enzymology , Stereoisomerism , Substrate Specificity
16.
DICP ; 25(7-8): 796-804, 1991.
Article in English | MEDLINE | ID: mdl-1835221

ABSTRACT

Goserelin acetate implant is a newly approved depot formulation of a luteinizing hormone-releasing hormone (LHRH) agonist indicated for palliation of advanced prostate cancer. LHRH superagonists suppress gonadotropin release from the pituitary gland by causing down-regulation of receptors. The sustained-release dosage form contains goserelin acetate dispersed in a biodegradable copolymer matrix and is designed to release active drug over 28 days. Pharmacokinetic studies have demonstrated that, despite nonzero order release of goserelin from the matrix, goserelin acetate implant maintains serum concentrations of testosterone in the range normally found in castrated men (less than 2 nmol/L) throughout the recommended 28-day dosing interval. Response rates similar to those for orchiectomy and estrogen administration have been demonstrated. Combination therapy with either diethylstilbestrol or flutamide has produced favorable results, although the major advantage appears to be a reduction in the tumor flare seen during the first week of LHRH agonist therapy rather than an increase in response rate or survival. Adverse effects are similar to other LHRH agonists and include tumor flare during the first week of therapy, decreased libido, decreased erectile potency, hot flashes, and gynecomastia. In combination with flutamide, additional adverse effects include diarrhea, nausea, vomiting, and elevated hepatic aminotransferases, all of which can be attributed to flutamide administration. Local reactions are minimal; however, some patients require a local anesthetic before goserelin acetate implant injection. The recommended dose is 3.6 mg administered subcutaneously into the upper abdominal wall every 28 days. The average wholesale cost is approximately +320 per month. Formulary addition is recommended.


Subject(s)
Buserelin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Amino Acid Sequence , Buserelin/administration & dosage , Buserelin/chemistry , Buserelin/pharmacokinetics , Buserelin/therapeutic use , Delayed-Action Preparations , Drug Implants , Goserelin , Humans , Male , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...