GB2416138A - Chromatographic separation of trilostane mixture - Google Patents

Chromatographic separation of trilostane mixture Download PDF

Info

Publication number
GB2416138A
GB2416138A GB0415830A GB0415830A GB2416138A GB 2416138 A GB2416138 A GB 2416138A GB 0415830 A GB0415830 A GB 0415830A GB 0415830 A GB0415830 A GB 0415830A GB 2416138 A GB2416138 A GB 2416138A
Authority
GB
United Kingdom
Prior art keywords
accordance
trilostane
chromatographic separation
approximately
mobile phase
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
GB0415830A
Other versions
GB0415830D0 (en
Inventor
Kevin Alexander Walker
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Stegram Pharmaceuticals Ltd
Original Assignee
Stegram Pharmaceuticals Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Stegram Pharmaceuticals Ltd filed Critical Stegram Pharmaceuticals Ltd
Priority to GB0415830A priority Critical patent/GB2416138A/en
Priority to PCT/GB2004/003348 priority patent/WO2006008430A1/en
Publication of GB0415830D0 publication Critical patent/GB0415830D0/en
Publication of GB2416138A publication Critical patent/GB2416138A/en
Withdrawn legal-status Critical Current

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/281Sorbents specially adapted for preparative, analytical or investigative chromatography
    • B01J20/282Porous sorbents
    • B01J20/285Porous sorbents based on polymers
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/22Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising organic material
    • B01J20/26Synthetic macromolecular compounds
    • B01J20/265Synthetic macromolecular compounds modified or post-treated polymers
    • B01J20/267Cross-linked polymers
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/42Selective adsorption, e.g. chromatography characterised by the development mode, e.g. by displacement or by elution
    • B01D15/424Elution mode
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2220/00Aspects relating to sorbent materials
    • B01J2220/50Aspects relating to the use of sorbent or filter aid materials
    • B01J2220/54Sorbents specially adapted for analytical or investigative chromatography

Landscapes

  • Chemical & Material Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Solid-Sorbent Or Filter-Aiding Compositions (AREA)
  • Treatment Of Liquids With Adsorbents In General (AREA)

Abstract

A method of determining Trilostane in a compound mixture comprising effecting a chromatographic separation of said mixture and analysing the eluate from the separation to determine Trilostane. The method characterised in that the chromatographic separation is effected using a chromatographic stationary phase comprised of a cross-linked resin incorporating aromatic groups. Preferably the chromatographic separation is effected in the presence of an aqueous mobile phase with a pH in the range 4-10. The preferred resin is a cross-linked styrene-divinylbenzene resin.

Description

METHOD FOR THE DETERMINATION OF TRlLOSTANE The present invention relates
to a method for the determination of Trilostane. In particular, but not exclusively, the invention relates to a method for the determination of Trilostane in a mixture containing Trilostane and structurally-related impurities formed during the preparation of Trilostane.
Trilostane ((4a,5a, 1 7,B)-4,5-epoxy-3,17-dihydroxyandrost-2-ene-2carbonitrile; 1) is used in human and veterinary medicine to treat endocrine disorders. Its primary indication is as an anticancer agent but it is also used in the treatment of Cushing's disease and other hyperadrenocortical conditions. :
HO (1)
TLC and HPLC have been used to investigate Trilostane and structurallyrelated compounds. For example, Mori et al.' describe the use of silica gels in preparative TLC methods for the separation and purification of Trilostane metabolites. Powles et al.2 and Robinson et al.3 disclose methods for the determination of Trilostane and a metabolite of Trilostane (the 17-keto analogue of Trilostane) in human plasma by HPLC employing a Hypersil octadecyl-substituted silica (ODS) column. Powles et al.2 also carried out TLC analysis of human plasma containing Trilostane using TLC plates coated with a silica gel.
Brown et al.4 describe an HPLC method for the determination of Trilostane and its 1 7-keto analogue using a silica-based Partisil column and a silica-based Phenyl/Corasil precolumn.
A study of Trilostane and ketotrilostane in rat plasma is described by McGee et al.5 involving the use of HPLC employing an ODS column.
In addition to use in academic and clinical investigations, silica-based stationary phases are the only type of stationary phase employed in the analysis of compound mixtures obtained during the industrial preparation of Trilostane.
The applicant has however unexpectedly found that the use of a silica or silica-based stationary phase in TLC methods for the determination of Trilostane leads to degradation of the target compound (i.e. Trilostane). Furthermore, the results of HPLC analyses of mixtures containing Trilostane and structurally-related compounds indicate that a conventional silica or silica-based stationary phase does not always provide adequate separation of Trilostane from the other compounds present. Consequently, uncertainties surround the accuracy and reliability of previous methods used to analyse Trilostane- containing mixtures.
According to a first aspect of the present invention there is provided a method of determining Trilostane in a compound mixture comprising effecting a chromatographic separation of said mixture and analysing the eluate from the separation to determine Trilostane, characterised in that the chromatographic separation is effected using a chromatographic stationary phase comprised of a cross-linked resin incorporating aromatic groups.
Use of a stationary phase comprised of a cross-linked resin incorporating aromatic groups avoids the previously unidentified problems outlined above in relation to the use of a silica or silica-based stationary phase. In particular, using a stationary phase in accordance with the present invention ensures that Trilostane is not degraded during the chromatographic separation process. An unforeseen advantage of the present invention is that, whereas previous methods employed a relatively polar silica or silica-based stationary phase, a relatively non-polar stationary phase comprised of a cross-linked resin incorporating aromatic groups facilitates sufficient separation of Trilostane from further compounds (including structurally-related compounds) to allow the accurate and reliable determination of Trilostane.
A second aspect of the present invention provides a method of separating Trilostane in a compound mixture comprising effecting a chromatographic separation of said mixture using a chromatographic stationary phase comprised of a cross-linked resin incorporating aromatic groups.
It is envisaged that a range of different stationary phases comprised of a cross-linked resin incorporating aromatic groups may be used in the method of the invention. It is preferred that the resin comprises styrene moieties. More preferably the resin comprises divinylbenzene moieties. Yet more preferably the resin is a cross-linked styrene- divinylbenzene resin.
The chromatographic separation may be HPLC. In this case, a stationary phase resin having any suitable bead size may be used. For example, the resin may have a bead size of up to approximately 10 m, although it is preferred that the resin has a bead size of approximately 3,um or approximately 5 m. Furthermore, the resin may have a pore size of up to approximately 10 1lm, for example the pore size of the resin may be approximately 3 m, approximately 5 lam or approximately 7 m. When HPLC is used, the HPLC column may have any appropriate length and/or diameter. Particularly suitable columns have a length of less than around 300 mm. By way of example, the HPLC column may have a length of about 50 mm, about 150 or about 250 mm. Moreover, the diameter of the HPLC column may be in the range 1 - 5 mm, more preferably in the range 2 - 4.5 mm. For example, the HPLC column may have a diameter of approximately 2.1 or approximately 4.1 mm.
When the method of the invention employs HPLC, the chromatographic separation may be effected at any desirable flow rate to ensure satisfactory separation of Trilostane over a reasonable timescale. It is preferred that the chromatographic separation is effected at a flow rate of up to approximately 1.0 ml/min, more preferably approximately 0.7 ml/mint The chromatographic separation according to the present invention can be carried out at ambient temperature or at an elevated temperature. Preferably the chromatographic separation is effected at a temperature of up to approximately 35 C, more preferably approximately 28 C.
The compound mixture will typically be introduced onto the chromatographic stationary phase as a solution (e.g. in methanol) and be eluted under the influence of a suitable mobile phase, preferably an aqueous mobile phase. Preferably the mobile phase has a pH in the range 4 - 10, more preferably 7.5 - 9.0, and most preferably a pH of approximately 8.3.
Thus, the mobile phase may comprise a buffer to maintain the pH of the mobile phase at a desired level. A preferred buffer is an acetate salt, such as ammonium acetate. The composition of the mobile phase should be selected to optimise the separation process for a particular application. To this end, it is preferred that the mobile phase incorporates a water miscible polar organic liquid, e.g. acetonitrile. Under certain conditions acetonitrile may be the sole constituent of the mobile in which case the mobile phase consists of 100 vol. % acetonitrile. In a preferred embodiment of the invention, a gradient elusion method is employed in which the percentage concentration of at least some of the components of the mobile phase, e.g. ammonium acetate solution and/or acetonitrile, are varied during the chromatographic separation process. Preferably, when a mobile phase consisting of ammonium acetate solution and acetonitrile is used, the percentage concentration of acetonitrile in the mobile phase is increased from around 10 vol. % to around 100 vol. % during the chromatographic separation with the balance of the mobile phase being made up with an appropriate amount of ammonium acetate solution. Thus, in this case, as the percentage concentration of acetonitrile is increased from around 10 vol. % to around 100 vol. % the percentage concentration of ammonium acetate solution in the mobile phase is decreased from around 90 vol. % to around O vol. %.
The rate at which the percentage concentration of a particular component of the mobile phase is varied during the chromatographic separation depends on the composition of the mobile phase and the period of time over which the chromatographic separation is carried out. The rate is preferably constant but, in certain circumstances, it may be more preferable to employ a rate which varies during the separation. It is preferred that the rate at which the percentage concentration of acetonitrile in the mobile phase is increased during the chromatographic separation is less than 10 % per minute, more preferably the rate lies in the range 2 to 7 % per minute, and most preferably the rate is about 4.5 % per minute.
The chromatographic separation may be carried out over any suitable period of time provided it is sufficiently long to ensure adequate separation and/or determination of Trilostane and any other compounds of interest which may be present. While the appropriate period of time will be influenced by many different factors such as the physical and chemical nature of the stationary and mobile phases, the flow rate and the temperature, it is preferred that the separation is carried out over a period of less than approximately I hour, more preferably less than around 30 minutes and most preferably around 20 minutes.
Where it is desired to reuse a chromatographic stationary phase following a chromatographic separation, a cleaning and re-equilibration cycle may be employed. For example, once all of the compounds of interest have been eluted from the stationary phase, the mobile phase may be maintained at its final composition for a further period of time (e.g. a few minutes) to ensure any remaining unwanted compounds are eluted from the column. Once the stationary phase has been cleaned in this way, the composition of the mobile phase running over the stationary phase may be adjusted to its original starting composition to re- equilibrate the stationary phase ready for further use.
It is envisaged that the method of the present invention is especially suited to the determination of Trilostane and structurally-related impurities formed during the preparation of Trilostane, and/or for the separation of Trilostane from such impurities.
While the present invention is well suited for use in HPLC methods, it is anticipated that the concept of using a stationary phase comprising a cross-linked resin incorporating aromatic groups may be applied to any suitable chromatographic technique. Moreover, it will be evident to the skilled person that the present invention may be applicable to both analytical and preparative chromatographic techniques.
The invention is illustrated with reference to the following non-limiting Examples.
REFERENCE EXAMPLES
The following experiments were carried out to investigate the stability of Trilostane during chromatographic analysis on a silica-based stationary phase.
Two samples of Trilostane were analysed by silica-based TLC employing a chloroform: methanol: glacial acetic acid (94: 4: 2 % v/v) mobile phase. The expected spots due to Trilostane and known related impurities (due to the method of preparation of Trilostane employed) were observed on each TLC plate, however, a number of other spots were observed on each plate indicating the presence of various unknown impurities. When each sample was reanalysed a few days later, additional unidentified spots were observed indicating that Trilostane and/or other compounds on the silicabased stationary phase had degraded whilst resident on the silica-based stationary phase. The results of these experiments illustrate the unsuitability of silica-based TLC methods for the accurate and reliable determination of Trilostane and related impurities.
In order to determine whether Trilostane degrades whilst in contact with a silica-based TLC stationary phase, a two dimensional TLC experiment was performed. A first dimension TLC experiment was carried out using a chloroform: methanol: glacial acetic acid (94: 4: 2 % v/v) mobile phase. The expected pattern of spots was obtained including those relating to Trilostane, known impurities and unknown impurities. The TLC plate was then rotated by 90 and a second dimension TLC experiment carried out using the same conditions as the first dimension experiment. If Trilostane was not degrading on the silica- based stationary phase, the second dimension experiment should have produced a single Trilostane spot because all related impurities should already have been separated from Trilostane in the first dimension experiment. However, this was not what was observed.
Following the second dimension experiment, a series of new spots were observed which strongly indicated that Trilostane had degraded on the silica-based stationary phase.
EXAMPLE 1
In view of the problems described in the Reference Example in relation to the degradation of Trilostane on a silica-based stationary phase, an improved HPLC system, described below, was developed for the determination of Trilostane Preparation of Test Mixture Containing Trilostane and Compounds X and Z mg of Trilostane was accurately weighed into a 100 ml volumetric flask and 80 ml of methanol added to form an initial solution of Trilostane in methanol. 25 mg of a structurally- related compound X and 25 mg of a structurally-related compound Z were accurately weighed into separate 25 ml volumetric flasks, dissolved in and made up to volume (i.e. up to 25 ml) with methanol. 2 ml of the solutions containing compounds X and Z was pipetted into the 100 ml flask containing the initial Trilostane solution in methanol and made to volume with further methanol to provide a Test Mixture containing 1 mg/ml Trilostane spiked with 0.02 mg/ml compound X and 0.02 mg/ml compound Z. Preparation of Mobile Phase Eluant A - 0.770 g of ammonium acetate was accurately weighed into a 1 1itre mobile phase jar along with I litre of deionised water. The pH of the solution was adjusted to pH 8.3 using ammonia solution (0.88 Sp. Gr) then sonicated prior to use. This provided a 10 mM solution of ammonium acetate.
Eluant B - 1 1itre of HPLC grade acetonitrile (1 M), which was sonicated prior to use.
HPLC Conditions Stationary Phase: styrene-divinylbenzene copolymer having a bead size of 3 1lm Mobile phase: ammonium acetate / acetonitrile, gradient as in Table 1 Flow rate: 0.7 ml/min Temperature: 28 C Injection volume: 10 al DAD* wavelength: 280 nm for Trilostane and compound X, 238 nm for compound Z *Diode Array Detection.
Mobile Phase Gradient Time (min) % Eluant A % Eluant B 0.0 90.0 10.0 20.0 0.0 100.0 25.0 0.0 100.0 25.1 90.0 10.0 30.0 90.0 l O.O
Table I
Between 0.0 and 20.0 minutes the concentration of Eluant A was decreased from 90.0 % to 0.0 % and the concentration of Eluant B was increased from 10.0 % to 100. 0 % at a controlled rate of 4.5 % per minute. Between 20.0 and 25.0 minutes the column was washed with a mobile phase consisting of 100.0 % Eluant B. Between 25.1 and 30.0 minutes the stationary phase was equilibrated with the use of a mobile consisting of 90.0 % Eluant A and 10.0 % Eluant B. HPLC analysis of the test mixture containing Trilostane and compounds X and Z was carried out using the above HPLC conditions. The DAD results obtained were used to produce a chromatogram, shown in Figure 1, and to calculate the results presented in Table 2.
Compound Retention Time Trilostane 8.88 X 9.67 Z 17.34
Table 2
The results presented in Figure 1 and Table 2 illustrate that the above HPLC system employing an aromatic cross-linked resin stationary phase does not cause degradation of Trilostane (or compounds X and Z) and facilitates good separation of Trilostane from structurally-related compounds X and Z. The method of the present invention therefore provides a robust, accurate and reliable method for the determination of Trilostane in a mixture containing Trilostane and at least one further compound.
EXAMPLE 2
The following experiments were carried out to compare two different HPLC systems for the analysis of a standard mixture containing Trilostane, the two structurally-related X and Z employed in Example 1 and a further structurally-related compound Y. A first system employed a silica-based stationary phase and a second system employed a stationary phase comprised of a cross-linked resin incorporating aromatic groups in accordance with the present invention. Two experiments were carried out using the second system to determine the reliability of the inventive method.
Preparation of Test Mixture Containing Trilostane and Compounds X, Y and Z The test mixture used in Example 2 was prepared in accordance with the procedure described in Example 1 for the preparation of the test mixture containing Trilostane and compounds X and Z save for appropriate modification such that the test mixture was spiked with 0.02 mg/ml of compounds X, Y and Z. Silica-Based Stationary Phase HPLC Conditions Stationary Phase: octadecyl substituted silica having a bead size of 5 rum Mobile Phase: water: methanol: acetonitrile: phosphoric acid (45:23:31:1 % v/v), isocratic Flow rate: 2.0 ml/min Temperature: 40 C Injection volume: 10 soul DAD wavelength: 254 rim HPLC analysis of the standard mixture containing Trilostane and compounds X, Y and Z was carried out using the above HPLC conditions. The DAD results obtained were used to produce a chromatogram, shown in Figure 2, and to provide the results presented in Table 3.
Compound Retention Time Trilostane 2.11
X 2.57 3.63
Table 3
The results presented in Figure 2 and Table 3 illustrate that a unique peak for compound X cannot be resolved from the predominant Trilostane peak. This demonstrates that the HPLC system employing a silica-based stationary phase does not provide adequate separation of compound X from Trilostane. This method is therefore not suitable for the determination of Trilostane and the structurally-related compounds X, Y and Z in a mixture of the four compounds.
Aromatic Cross-linked Resin Stationary Phase Preparation of Mobile Phase An ammonium acetate (10 mM, pH 8.3) / acetonitrile mobile phase was prepared in accordance with the procedure set out in Example 1.
HPLC Conditions Stationary phase: styrene-divinylbenzene copolymer having a bead size of 3 Em Mobile phase: ammonium acetate / acetonitrile, gradient as in Example 1, Table 1 Flow rate: 0.7 ml/min Temperature: 28 C Injection volume: 10 Ill DAD wavelength: 280 rim for Trilostane and compound X, 244 for compound Y and 238 rim for compound Z Two separate HPLC analyses of the test mixture containing Trilostane and compounds X, Y and Z were carried out using the above HPLC conditions. The DAD results obtained were used to produce two chromatograms (Run 1: Figure 3 and Run 2: Figure 4) and to provide the results presented in Table 4.
Compound Retention Time Retention Time (Run 1) (Run 2) Trilostane 8.76 9.10 X 9.52 9.92 Y 15.41 15.53 17.16 17.18
Table 4
The results presented in Figures 3 and 4, and Table 4 show that the HPLC system employing an aromatic cross-linked resin stationary phase does not cause degradation of Trilostane (or compounds X, Y and Z) and facilitates good separation of Trilostane from structurally-related compounds X, Y and Z. Thus, the HPLC method of the present invention provides an accurate and reliable method for the determination of Trilostane and represents a significant advance over existing methods, which have now been shown to be unreliable and potentially inaccurate.
REFERENCES
1. Mori Y., Makoto T. and Makoto S., Chem. Pharm. Burt., 1981, 29(9), 2478-84.
2. Powles P., Robinson D.T., Andrews R.S. and Robinson P.R., J. Chromatogr., 1984, 311(2), 434-42.
3. Robinson D.T., Earnshaw R.J., Mitchell R., Powles P., Andrews R.S. and Robertson W.R., J. Steroid Biochem., 1984, 21(5), 601-5.
4. Brown, Stroshane and Benziger, J. Chromatogr., 1985, 339, 440-44.
5. McGee, Palin and Shaw, J. Chromatogr., 1991, 567, 282-7. t

Claims (24)

1. A method of determining Trilostane in a compound mixture comprising effecting a chromatographic separation of said mixture and analysing the eluate from the separation to determine Trilostane, characterized in that the chromatographic separation is effected using a chromatographic stationary phase comprised of a cross-linked resin incorporating aromatic groups.
2. A method of separating Trilostane in a compound mixture comprising effecting a chromatographic separation of said mixture using a chromatographic stationary phase comprised of a cross-linked resin incorporating aromatic groups.
3. A method in accordance with claim 1 or 2, wherein the resin comprises styrene moieties.
4. A method in accordance with claim 1, 2 or 3, wherein the resin comprises divinylbenzene moieties.
5. A method in accordance with any one of claims 1 to 4, wherein the resin is a cross- linked styrene-divinylbenzene resin.
6. A method in accordance with any preceding claim, wherein the resin has a bead size of up to approximately 10 m.
7. A method in accordance with any one of claims 1 to 5, wherein the resin has a bead size of approximately 3 1lm or approximately 5 m.
8. A method in accordance with any preceding claim, wherein the chromatographic separation is effected at a flow rate of up to approximately 1.0 ml/mint
9. A method in accordance with any one of claims l to 7, wherein the chromatographic separation is effected at a flow rate of approximately 0. 7 ml/mint
l O. A method in accordance with any preceding claim, wherein the chromatographic separation is effected at a temperature of up to approximately 35 C.
11. A method in accordance with any one of claims 1 to 9, wherein the chromatographic separation is effected at a temperature of approximately 28 C.
12. A method in accordance with any preceding claim, wherein the chromatographic separation is carried out over a period of less than approximately 1 hour.
13. A method in accordance with any one of claims 1 to 11, wherein the chromatographic separation is carried out over a period of less than around 30 minutes.
14. A method in accordance with any one of claims 1 to 11, wherein the chromatographic separation is carried out over a period of less than around 20 minutes.
15. A method in accordance with any preceding claim, wherein the chromatographic separation is effected in the presence of an aqueous mobile phase.
16. A method in accordance with claim 15, wherein the mobile phase has a pH in the range 4 - 10.
17. A method in accordance with claim 15, wherein the mobile phase has a pH in the range 7.5 - 9.0.
18. A method in accordance with claim 15, wherein the mobile phase has a pH of approximately 8.3.
l 9. A method in accordance with any one of claims 15 to 18, wherein the mobile phase comprises a buffer to maintain the pH of the mobile phase at a desired level.
20. A method in accordance with claim 19, wherein the buffer is an acetate salt.
21. A method in accordance with claim 20, wherein the acetate salt is ammonium acetate.
22. A method in accordance with any one of claims 15 to 21, wherein the mobile phase incorporates an organic liquid.
23. A method in accordance with claim 22, wherein the organic liquid is acetonitrile.
24. A method in accordance with claim 23, wherein the mobile phase incorporates up to approximately 100 vol. % acetonitrile.
GB0415830A 2004-07-15 2004-07-15 Chromatographic separation of trilostane mixture Withdrawn GB2416138A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
GB0415830A GB2416138A (en) 2004-07-15 2004-07-15 Chromatographic separation of trilostane mixture
PCT/GB2004/003348 WO2006008430A1 (en) 2004-07-15 2004-08-04 Method for the determination of trilostane

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB0415830A GB2416138A (en) 2004-07-15 2004-07-15 Chromatographic separation of trilostane mixture

Publications (2)

Publication Number Publication Date
GB0415830D0 GB0415830D0 (en) 2004-08-18
GB2416138A true GB2416138A (en) 2006-01-18

Family

ID=32893600

Family Applications (1)

Application Number Title Priority Date Filing Date
GB0415830A Withdrawn GB2416138A (en) 2004-07-15 2004-07-15 Chromatographic separation of trilostane mixture

Country Status (2)

Country Link
GB (1) GB2416138A (en)
WO (1) WO2006008430A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1129125A (en) * 1965-02-24 1968-10-02 Rohm & Haas Enrichment and/or separation of an organic compound by adsorption processes
JPS5175074A (en) * 1974-12-18 1976-06-29 Nippon Kemikaru Kk Tokofuerooruno seiseinoshukuho
JPS63166899A (en) * 1986-12-29 1988-07-11 Tosoh Corp Separation and determination of steroid hormone
JPH01224396A (en) * 1988-03-01 1989-09-07 Wakunaga Pharmaceut Co Ltd Novel steroid saponin and production and novel use of these derivative

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8404980D0 (en) * 1984-02-25 1984-03-28 Sterwin Ag Steroid compound
JP2001261695A (en) * 2000-03-14 2001-09-26 Tosoh Corp Method and apparatus for concentrating steroids

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1129125A (en) * 1965-02-24 1968-10-02 Rohm & Haas Enrichment and/or separation of an organic compound by adsorption processes
JPS5175074A (en) * 1974-12-18 1976-06-29 Nippon Kemikaru Kk Tokofuerooruno seiseinoshukuho
JPS63166899A (en) * 1986-12-29 1988-07-11 Tosoh Corp Separation and determination of steroid hormone
JPH01224396A (en) * 1988-03-01 1989-09-07 Wakunaga Pharmaceut Co Ltd Novel steroid saponin and production and novel use of these derivative

Also Published As

Publication number Publication date
GB0415830D0 (en) 2004-08-18
WO2006008430A1 (en) 2006-01-26

Similar Documents

Publication Publication Date Title
Li et al. Simultaneous determination of clenbuterol, salbutamol and ractopamine in milk by reversed-phase liquid chromatography tandem mass spectrometry with isotope dilution
Ravanat et al. Isotope dilution high-performance liquid chromatography–electrospray tandem mass spectrometry assay for the measurement of 8-oxo-7, 8-dihydro-2′-deoxyguanosine in biological samples
Gracia-Lor et al. Simultaneous determination of acidic, neutral and basic pharmaceuticals in urban wastewater by ultra high-pressure liquid chromatography-tandem mass spectrometry
Noche et al. Microextraction by packed sorbent for the analysis of pharmaceutical residues in environmental water samples by in situ derivatization-programmed temperature vaporizer–gas chromatography–mass spectrometry
de Castro et al. High-throughput on-line solid-phase extraction–liquid chromatography–tandem mass spectrometry method for the simultaneous analysis of 14 antidepressants and their metabolites in plasma
Wang et al. Development of a chiral micellar electrokinetic chromatography–tandem mass spectrometry assay for simultaneous analysis of warfarin and hydroxywarfarin metabolites: Application to the analysis of patients serum samples
Trontelj Quantification of glucuronide metabolites in biological matrices by LC-MS/MS
Song et al. Analysis of omeprazole and 5-OH omeprazole in human plasma using hydrophilic interaction chromatography with tandem mass spectrometry (HILIC–MS/MS)—Eliminating evaporation and reconstitution steps in 96-well liquid/liquid extraction
Nemoto et al. High-throughput determination of nonsteroidal anti-inflammatory drugs in human plasma by HILIC-MS/MS
Petruczynik et al. Determination of some psychotropic drugs in serum and saliva samples by HPLC-DAD and HPLC MS
Naidong et al. Development and validation of a hydrophilic interaction liquid chromatography–tandem mass spectrometric method for the analysis of paroxetine in human plasma
Ferreira et al. In situ derivatization coupled to microextraction by packed sorbent and gas chromatography for the automated determination of haloacetic acids in chlorinated water
Li et al. Enantiomeric separation and enantioselective determination of some representive non-steroidal anti-inflammatory drug enantiomers in fish tissues by using chiral liquid chromatography coupled with tandem mass spectrometry
Miwa et al. High-performance liquid chromatographic analysis of serum short-chain fatty acids by direct derivatization
Wróblewski et al. Determination of vortioxetine in human serum and saliva samples by HPLC–DAD and HPLC–MS
Clauson-Kaas et al. UPLC-MS/MS determination of ptaquiloside and pterosin B in preserved natural water
Chen et al. Determination of ten haloacetic acids in drinking water using high-performance and ultra-performance liquid chromatography-tandem mass spectrometry
Fiori et al. Evaluation of two different clean-up steps, to minimise ion suppression phenomena in ion trap liquid chromatography–tandem mass spectrometry for the multi-residue analysis of beta agonists in calves urine
Olesen et al. Determination of nortriptyline in human serum by fully automated solid-phase extraction and on-line high-performance liquid chromatography in the presence of antipsychotic drugs
Yang et al. Simultaneous quantitation of the diastereoisomers of scholarisine and 19‐epischolarisine, vallesamine, and picrinine in rat plasma by supercritical fluid chromatography with tandem mass spectrometry and its application to a pharmacokinetic study
Tuzimski et al. A QuEChERS-based sample preparation method for the analysis of 5-nitroimidazoles in bovine milk by HPLC–DAD
Umezawa et al. Simultaneous determination of β‐blockers in human plasma using liquid chromatography–tandem mass spectrometry
Nobilis et al. High-performance liquid chromatographic determination of tiapride and its phase I metabolite in blood plasma using tandem UV photodiode-array and fluorescence detection
Xiong et al. Determination of co-administrated opioids and benzodiazepines in urine using column-switching solid-phase extraction and liquid chromatography–tandem mass spectrometry
Li et al. Enantioselective determination of ketamine in dog plasma by chiral liquid chromatography–tandem mass spectrometry

Legal Events

Date Code Title Description
WAP Application withdrawn, taken to be withdrawn or refused ** after publication under section 16(1)