1 / 29

Multi-Analyte LC-MS/MS Methods – Best Practice.

Multi-Analyte LC-MS/MS Methods – Best Practice. Martin Danaher. Contents. LC-MS/MS Overview LC-MS/MS Optimisation Method Validation Specificity and selectivity Stability studies WLr and WLR Data quality checks Conclusions. LC-MS/MS Overview. Sample manager. Column oven Injector.

lreyna
Télécharger la présentation

Multi-Analyte LC-MS/MS Methods – Best Practice.

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Multi-Analyte LC-MS/MS Methods – Best Practice. Martin Danaher

  2. Contents • LC-MS/MS Overview • LC-MS/MS Optimisation • Method Validation • Specificity and selectivity • Stability studies • WLr and WLR • Data quality checks • Conclusions

  3. LC-MS/MS Overview Sample manager Column oven Injector Pumps MS/MS (QqQ) 3 3

  4. LC-MS/MS instrumentation (HT) Column manager: 4 columns Sample organizer: 10 trays Injector : 48 positions

  5. Electrospray Ionisation

  6. MS Optimisation • MS Optimisation • Rafoxanide: EF = C19H11Cl2I2NO3 • MW: 626.01 g/mol (average value) • Monoisotopic 624.820496 • Which polarity? • Optimum cone voltages • ESI voltage • Temperatures

  7. Information sources http://www.sisweb.com/mstools/isotope.htm

  8. Isotopic Distribution of Rafoxanide

  9. MS/MS Optimisation • Collision induced dissociation with inert gas e.g. N2 or Argon. • Identify most abundant?? daughter or product ions • Product ions must be selective • Avoid neutral losses -18 (H2O) and -17 (OH) • Avoid non-specific fragments: 91 m/z, 105 m/z and 121 m/z. • Consult literature, see what others are using.

  10. Chromatographic development • Generic scouting gradient • M. Phase A 100% Aqueous • M. Phase B Acetonitrile or Methanol • Column: 100 × 2.1 mm • Inject each mix at high concentration and optimise separation • Evaluate the impact of different additives acids and salts • Optimise additive concentrations

  11. Start method validation

  12. 2002/657/EC Criteria

  13. Specificity – Similar compounds • Inject analyte standard and internal standard separately (highest concentration). • Check for interference in each analyte or IS trace • Isobaric interference • Cross-talk • Carry-over

  14. Isobaric interference

  15. Cross-talk phenomenon

  16. Isotopic Distribution of Rafoxanide

  17. Selectivity – Matrix components • HPLC-FLD separation of analyte from the matrix peak.

  18. Selectivity – Matrix components • LC-MS/MS – Matrix peaks not visible • Co-eluting peaks, late eluting peaks, etc. • Ion suppression or enhancement • Potential Solutions: • Clean-up • Chromatographic separation • Matrix matched standards • SILs

  19. Matrix Effects Study – Approach I • Post-column infusion of standards with blank matrix samples

  20. Matrix Effects Study Example

  21. Matrix Effects Study – Approach II • Spike a range of representative samples post extraction and compare with solvent standards. • Calculate enhancement or suppression effects • Calculate the precision • Evaluate the impact of the use of internal standards

  22. Importance of Chromatography

  23. Importance of Chromatography

  24. Method Validation • Stability studies • Standard stability (3, 6, 12, 24, 36 months). Different storage conditions. • Sample extracts – intermediate or in final injection solvent. Over 7 days or continuous injection • Stability in matrix – spike samples and store for different periods of time (1, 2, 3, 4, 6, 8, 12, 26, 52 weeks) • Limit of detection • Limit of quantitation/Limit of Reporting

  25. Method Validation • Within laboratory repeatability • 18 samples spiked at three different levels • Repeat by the same analyst • Within laboratory reproducibility • Minimum of 18 “different” samples spike at three different levels • Repeat on different days by the different analysts. • Use different equipment if possible. • CCα • Calculate using WLR data.

  26. Data quality Checks (qualitative) • Identification • RT (5%)/RRT (2.5%) • S/N >3 • Identification points (3 or 4) • Ion ratio

  27. Data quality Checks (quantitative) • CCα • Compare CCα with MRL. Big gap  more precise method needed. • Calibrations • Use weighted linear regression not through (0,0) • Inject at start and end of batch. Drift <30%. • Inject LOQ as a response check throughout the run. Drift <30%. • Residuals ±20%. • Minimum of five points on curve.

  28. Data quality Checks (quantitative) • Trueness • Precision For analyses carried out under repeatability conditions, the intra-laboratory CV would typically be between ½ and 2/3 of the above values.

  29. Conclusions • SANCO validation document presents complementary validation guidelines. • Provides more practical information on routine analysis • Interpretation of data quality. • Elements in 2002/657/EC validation that should be retained. • Good ideas e.g. Ccα • Ambiguity around the validation approach • Considered as being inflexible.

More Related