article

A process analytical tool

Posted: 7 March 2005 |

There is an increasing demand for new approaches to understand the chemical and physical phenomena that occur during pharmaceutical unit operations. Obtaining real-time information from processes opens new perspectives for safer manufacture of pharmaceuticals. Raman spectroscopy provides a molecular level insight into processing and it is therefore a promising process analytical tool.

There is an increasing demand for new approaches to understand the chemical and physical phenomena that occur during pharmaceutical unit operations. Obtaining real-time information from processes opens new perspectives for safer manufacture of pharmaceuticals. Raman spectroscopy provides a molecular level insight into processing and it is therefore a promising process analytical tool.

There is an increasing demand for new approaches to understand the chemical and physical phenomena that occur during pharmaceutical unit operations. Obtaining real-time information from processes opens new perspectives for safer manufacture of pharmaceuticals. Raman spectroscopy provides a molecular level insight into processing and it is therefore a promising process analytical tool.

Gathering relevant information from multicomponent systems, such as pharmaceutical unit operations, is not a straightforward task. Consider a typical solid dosage form with numerous sequential processing steps. There are many possible pitfalls during processing that may critically affect the final product performance. For example, active pharmaceutical ingredient or excipient may be stressed in an aqueous environment or they may be stressed thermally during processing. Focusing analysis on the end product will not enable the early detection of problems or further, the complex relations between them. Recently, the U.S. FDA introduced a guidance to address this issue1.

Process analytical technology (PAT) is a system for developing and implementing new efficient tools for use during pharmaceutical development, manufacturing and quality assurance, while maintaining or improving the current level of product quality assurance. This guideline categorises PAT tools in five groups: multivariate tools for design; data acquisition and analysis; process analysers; process control tools and continuous improvement and knowledge management tools. Raman spectroscopy provides a molecular level insight into processing and therefore offers a new way to understand our unit operations. The basic principle of Raman spectroscopy is to irradiate a substance with monochromatic light and to detect the scattered light with different wavelength to the incident beam. The differences in the frequencies result in characteristic Raman shifts. The Raman effect is inherently very weak and, in addition to intense excitation source, good filters are needed to remove the excitation line from the collected radiation. Utilisation of this phenomenon has been relatively limited in the field of pharmaceutical processing due to the high price of instrumentation and difficulties in process interfacing. Recent developments in the fields of optoelectronics, computer technology, data transfer and data analysis methods have enabled the real-time and non-invasive Raman analysis of pharmaceutical unit operations and, by this means, a molecular level insight into processing. This will enable process understanding for scientific, risk-managed pharmaceutical development, manufacture and quality assurance in accordance with the PAT ideology.

Raman spectroscopy within pharmaceutical unit operations

There are an increasing number of published studies on the utilisation of Raman spectroscopy in the process environment. Vankeirsbilck et al. have recently reviewed the use of Raman in the field of pharmaceutics, with attention paid to comparison of FT-Raman and dispersive instruments2. Threlfall3 and Bugay4 have reviewed the use of spectroscopic tools for solid-state analysis and in these reviews they relate Raman to the other tools available. Issues related to quantitative analysis with Raman are well described in a tutorial by Pelletier5. This chapter summarises the possibilities of Raman spectroscopy to the process analysis of pharmaceutical unit operations related to solid dosage forms. Discussion starts from the synthesis phase and concludes with the film coating process. Svensson et al. used Raman spectroscopy for reaction monitoring in combination with multivariate techniques6. To avoid problems related to spectral overlapping, they recommend the use of effective preprocessing (standard normal variate and derivatives) together with principal component analysis (PCA) and partial least squares (PLS). They achieved a rate constant with a model system, with good agreement with published values. The subsequent processing step is crystallization of material. This critical unit operation is performed to produce purified material with desired purity, polymorphic composition, surface properties and particle size and shape distributions. It is crucial to have an in-depth process signature from crystallization phase, as a failure in crystallization results in major difficulties in secondary manufacturing steps (mixing, granulation, tableting, coating). Crystallization is not a well understood nor controlled unit operation. The recent case of ritonavir really underlines the need for new tools in the process analysis and control of crystallization and in the implementation of a polymorph screening step7. However, the number of published works on real-time analysis of crystallization with pharmaceutics is relatively limited. Schwartz monitored in situ lysozyme concentration changes in hanging drop crystallization8. Changes in polymorphic composition has been monitored and quantified with in-line Raman spectroscopy9-14. Falcon and Berglund reported the use of Raman for real time monitoring of phenomena related to antisolvent addition13. Recently, Hu et al. reported simultaneous monitoring of solution concentration in conjunction with information about the polymorphic outcome of the crystallization event and further, solvent-mediated transformations of the model system14. Raman spectroscopy can also be used to understand the mechanisms of the phase transitions15. Batch crystallizations of pharmaceutics are quite often performed in aqueous media, so Raman spectroscopy is an extremely promising tool for process control and monitoring purposes. In the solid state quantification of polymorphic form, Raman spectroscopy is an ideal candidate. Minimal sample preparation combined with sensitivity to polymorphism opens new perspectives for fast and reliable solid state analysis16-22. Both univariate and multivariate methods have been used for development of quantitative models. In addition, the use of Raman for quantification of crystallinity has been reported23. Recently, Raman has been combined with high-throughput (HTS) polymorph screening ideology 24,25. There is an increasing demand for early screening of solid-state forms and further identification of the most stable form. After a case related to polymorphism of ritonavir, high-throughput crystallization experiments were carried out to explore the diversity of ritonavir solid state forms26. One of the least understood unit operations within solid dosage forms is the mixing of powders. Vergote et al. have reported the use of Raman for in-line monitoring of the blending process27. Raman mapping in combination with near IR spectral mapping can be used to describe heterogeneous mixtures in more detail28. The granulation step – and in some cases wet granulation – is a process step needed for many products. In this unit operation, material may undergo phase transformation after exposure to solvent29. Possible phase transitions are polymorphic transformations; solvate formation and dehydration of solvate; production of amorphous regions and crystallization of amorphous material. The use of Raman for at-line30 and in-line31 analysis of hydrate formation during wet granulation has been reported. Wikström et al. used the real time information to verify a model for predicting the transformation kinetics of hydrate formation. Raman spectroscopy also provides an insight into water-solid interactions in the formulation and further can be used to understand the role of excipients in the early development phase. Taylor et al. investigated the nature of water-polymer interactions for polymers of pharmaceutical interest32. Airaksinen et al. has reported the use of Raman to detect hydrate formation in presence of excipients and the role of excipients in the phase transformation33. FT-Raman has been utilised in evaluating the potential of carrageens to protect drugs from polymorphic transformations34. Schmidt et al. reported the detection of both recrystallization of amorphous component and dehydration after the tabletting process. Fechner et al. utilised Raman in the extrusion-spheronization process environment35. They explained the effect of water on the structure of cellulose during this unit operation. One of the most attractive possibilities of Raman and other possible PAT sensors is to utilise them for the real time assay of tablets and capsules. Moving into a situation where we can analyse, for example, every tenth tablet during production, will open up totally new perspectives for quality assurance. Raman has been used for quantification of components in antacid tablets36. Wang et al. reported the use of Raman for direct assay of acetylsalicylic acid and also the analysis of the major degradation product, salicylic acid37. Niemczyk et al. utilised this technique for quantitative analysis of intact gel capsules and they also reported the analysis of capsules through blister packs38. Vergote et al. investigated the role of excipients in quantification of diltiazem hydrochloride39. Folestad and Johansson have recently discussed the use of Raman for monitoring the tableting process40. One application area for Raman is the fast analysis of prohibited substances from seized tablets, as reported by Bell et al. for analysis of ecstasy (MDMA, N-methyl-3,4-methylenedioxyamphetamine and its near analogues). Another potential consideration is the use of Raman for fast analysis of possible processing-induced transformation during the tableting process. It could also be used for fast verification of the polymorphic form of a drug in final tablets21,42. Again, solid state properties of both excipients and active pharmaceutical ingredients can be followed non-invasively. In many cases, following unit operation is the coating process, usually performed using an aqueous polymer solution. Raman spectroscopy has been utilised in various other areas for analysis of film coatings, but not widely in the field of pharmaceutics. Ringqvist et al. has reported the use of confocal Raman for analysis of the chemical composition in selected small areas of the coating surface43.

Challenges in process analysis

One fundamental question concerning process measurements with Raman is interfacing into process, as it is with all process analytical tools. The basic problem is obviously to keep the insight into a process clean, to measure a representative part of the material and to have the moving sample in focus. A Raman probe can be mounted invasively using an immersion probe, or process monitoring can be performed non-invasively using non-contact optics. Another problem related to Raman is the small sampling area. The penetration depth of the lasers used is relatively small, resulting in a small effective sample volume. This can be affected with optics, by increasing the area that is being measured. Wikström et al. has recently evaluated different sampling devices for in-line measurements44. Sample heating is a widely recognised problem in Raman spectroscopy. Moving the sample being measured, which is automatically the case in process analysis, can minimise problems related to heating. Johansson et al. investigated the sample heating of pharmaceutical materials and developed a model to predict the rotation speed needed to minimise the heating45. With some materials, an intense fluorescence background is observed. This can be affected by selecting the appropriate excitation wavelength.

Conclusions

Raman spectroscopy has matured into an effective tool for ensuring safe and efficient manufacturing of pharmaceutics. Much has happened since Chandrasekhara Venkata Raman visited Europe in the summer of 1921, when he formulated his first ideas related to this phenomena whilst observing the blue opalescence of the Mediterranean Sea46 At the moment, we have instruments ready for non-invasive process measurements. Recent developments in the fields of optoelectronics, computer technology, data transfer and data analysis methods have enabled the real-time and non-invasive Raman analysis of pharmaceutical unit operations and, by this means, a molecular level insight into processing. More research is needed to understand the full potential of Raman as a process analytical tool.

References

  1. PAT — a framework for innovative pharmaceutical development, manufacturing, and quality assurance In Guidance for industry; U.S. Food and Drug Administration: Rockville, MD, 2004.
  2. T. Vankeirsbilck, A. Vercauteren, W. Baeyens, G. Van der Weken, F. Verpoort, G. Vergote, J.P. Remon, 2002. Applications of Raman spectroscopy in pharmaceutical analysis. TrAC 21: 869-877.
  3. T. Threlfall, 1995. Analysis of organic polymorphs. Analyst 120: 2435-2460.
  4. D. Bugay, 2001. Characterization of the solid- state: spectroscopic techniques. Adv. Drug Del. Rev. 48: 43-65.
  5. M. Pelletier, 2003. Quantitative analysis using Raman spectrometry. Appl. Spectrosc. 57: 20A-41A.
  6. O. Svensson, M. Josefson, F. Langkilde, 1999. Reaction monitoring using Raman spectroscopy. Chem. Intell. Lab. Syst. 49: 49-66.
  7. J. Bauer, S. Spanton, R. Henry, J. Quick, W. Dziki, W. Porter, J. Morris, 2001. Ritonavir: an extraordinary example of conformational polymorphism. Pharm. Res. 18: 859-866.
  8. A. Schwartz, K. Berglund, 1999. The use of Raman spectroscopy for in situ monitoring of lysozyme concentration during crystallization in a hanging drop. J. Cryst. Growth 203: 599-603.
  9. F. Wang, J. Wachter, F. Antosz, K. Berglund, 2000. An investigation of solvent-mediated polymorphic transformation of progesterone using in situ Raman spectroscopy. Org. Proc. Res. Dev. 4: 391-395.
  10. C. Starbuck, A. Spartalis, L. Wai, J. Wang, P. Fernandez, C. Lindemann, G. Zhou, Z. Ge, 2002. Process optimization of a complex pharmaceutical polymorphic system via in situ Raman spectroscopy. Cryst. Growth Des. 2: 515-522.
  11. E. Ferrari, R. Davey, 2004. Solution-mediated transformation of _ to _ l-glutamic acid: rate enhancement due to secondary nucleation. Cryst. Growth Des. 4: 1061-1068.
  12. T. Ono, J. ter Horst, P. Jansens, 2004. Quantitative measurement of the polymorphic transformation of l-glutamic acid using in-situ Raman spectroscopy. Cryst. Growth Des. 4: 465-469.
  13. J. Falcon, K. Berglund, 2004. In situ monitoring of antisolvent addition crystallization with principal components analysis of Raman spectra. Cryst. Growth Des. 4: 457-463.
  14. Y. Hu, J. Liang, A. Myerson, L. Taylor, 2005. Crystallization monitoring by Raman spectroscopy: simultaneous measurement of desupersaturation profile and polymorphic form in flufenamic acid systems. Ind. Eng. Chem. Res. (ASAP article).
  15. S. Boerrigter, C. van den Hoogenhof, H. Meekes, P. Bennema, E. Vlieg, P. J. C. M. van Hoof, 2002. In situ observation of epitaxial polymorphic nucleation of the model steroid methyl analogue 17 norethindrone. J. Phys. Chem. B 106: 4725-4731.
  16. C. Deeley, R. Spragg, T. Threlfall, 1991. A comparison of Fourier transform infrared and near-infrared Fourier transform Raman spectroscopy for quantitative measurements: an application in polymorphism. Spectrochim. Acta 47A: 1217-1223.
  17. F. Langkilde, J. Sjöblom, L. Tekenbergs-Hjelte, J. Mrak, 1997. Quantitative FT-Raman analysis of two crystal forms of a pharmaceutical compound. J. Pharm. Biomed. Anal. 15: 687-696.
  18. P. Findlay, D. Bugay, 1998. Utilization of Fourier transform-Raman spectroscopy for the study of pharmaceutical crystal forms. J. Pharm. Biomed. Anal. 16: 921-930.
  19. S. Campbell Roberts, A Williams, I. Grimsey, S. Booth, 2002. Quantitative analysis of mannitol polymorphs. FT-Raman spectroscopy. J. Pharm. Biomed. Anal. 28: 1135-1147.
  20. N. Al-Zoubi, J. Koundourellis, S. Malamataris, 2002. FT-IR and Raman spectroscopic methods for identification and quantitation of orthorhombic and monoclinic paracetamol in powder mixes. J. Pharm. Biomed. Anal. 29: 459-467.
  21. M. Auer, U. Griesser, J. Sawatzki, 2003. Qualitative and quantitative study of polymorphic forms in drug formulations by near infrared FT-Raman spectroscopy. J. Molec. Struct. 661-662: 307-317.
  22. C. Strachan, D. Pratiwi, K. Gordon, T. Rades, 2004. Quantitative analysis of polymorphic mixtures of carbamazepine by Raman spectroscopy and principal component analysis. J. Raman Spectr. 35: 347-352.
  23. L. Taylor, G. Zografi, 1998. Quantitative analysis of crystallinity using FT-Raman spectroscopy. Pharm. Res. 15: 755-761.
  24. M. Peterson, S. Morissette, C. McNulty, A. Goldsweig, P. Shaw, M. LeQuesne, J. Monagle, N. Encina, J. Marchionna, A. Johnson, J. Gonzalez-Zugasti, A. Lemmo, S. Ellis, M. Cima, Ö. Almarsson, 2002. Iterative high-throughput polymorphism studies on acetaminophen and an experimentally derived structure for Form III. J. Am. Chem. Soc. 124: 10958-10959.
  25. C. Anderton, 2004. A valuable technique for polymorph screening. Eur. Pharm. Rev. 9(2): 68-74.
  26. S. Morissette, S. Soukasene, D. Levinson, M. Cima, Ö. Almarsson, 2003. Elucidation of crystal form diversity of the HIV protease inhibitor ritonavir by high-throughput crystallization. Proc. Natl. Acad. Sci USA. 100: 2180–2184.
  27. G. Vergote, T. De Beer, C. Vervaet, J.P. Remon, W. Baeyens, N. Diericx, F. Verpoort, 2004. In-line monitoring of a pharmaceutical blending processusing FT-Raman spectroscopy. Eur. J. Pharm. Sci. 21: 479-485.
  28. F. Clarke, M. Jamieson, D. Clark, S. Hammond, R. Jee, A. Moffat, 2001. Chemical image fusion. The synergy of FT-NIR and Raman mapping microscopy to enable a more complete visualization of pharmaceutical formulations. Anal. Chem. 73: 2213-2220.
  29. K. Morris, U. Griesser, C. Eckhardt, J. Stowell, 2001. Theoretical approaches to physical transformations of active pharmaceutical ingredients during manufacturing processes. Adv. Drug Deliver. Rev. 48: 91-114.
  30. A. Jørgensen, J. Rantanen, M. Karjalainen, L. Khriachtchev, E. Räsänen, J. Yliruusi, 2002. Hydrate formation during wet granulation studied by spectroscopic methods and multivariate analysis. Pharm. Res. 19: 1282-1288.
  31. H. Wikström, P. Marsac, L. Taylor, 2005. In-line monitoring of hydrate formation during wet granulation using Raman spectroscopy. J. Pharm. Sci. 94: 209-219.
  32. L. Taylor, F. Langkilde, G. Zografi, 2001. Fourier transform Raman spectroscopic study of the interaction of water vapor with amorphous polymers. J. Pharm. Sci. 90: 888-901.
  33. S. Airaksinen, P. Luukkonen, A. Jørgensen, M. Karjalainen, J. Rantanen, J. Yliruusi, 2003. Effects of excipients on hydrate formation in wet masses containing theophylline. J. Pharm. Sci. 92: 516-528.
  34. A. Schmidt, S. Wartewig, K. Picker, 2003. Potential of carrageenans to protect drugs from polymorphic transformation. Eur. J. Pharm. Biopharm. 56: 101-110.
  35. P. Fechner, S. Wartewig, M. Füting, A. Heilmann, R. Neubert, P. Kleinebudde, 2003. Properties of microcrystalline cellulose and powder cellulose after extrusion/spheronization as studied by Fourier transform Raman spectroscopy and environmental scanning electron microscopy. AAPS PharmSci 5: Article 31.
  36. C. Kontoyannis, 1995. Quantitative determination of CaCO3 and glycine in antacid tablets by laser Raman spectroscopy. J. Pharm. Biomed. Anal. 13: 73-76.
  37. C. Wang, T. Vickers, C. Mann, 1997. Direct assay and shelf-life monitoring of aspirin tablets using Raman spectroscopy. J. Pharm. Biomed. Anal. 16: 87-94.
  38. T. Niemczyk, M. Delgado-Lopez, F. Allen, 1998. Quantitative determination of bucindolol concentration in intact gel capsules using Raman spectroscopy. Anal. Chem. 70: 2762-2765.
  39. G. Vergote, C. Vervaet, J. P. Remon, T. Haemers, F. Verpoort. Near-infrared FT-Raman spectroscopy as a rapid analytical tool for the determination of diltiazem hydrochloride in tablets. Eur. J. Pharm. Sci. 16: 63-67.
  40. J. Johanson, S. Folestad, 2003. Raman Spectroscopy Opening the PAT toolbox. Eur. Pharm. Rev. 8(4): 36-42.
  41. S. Bell, D. Burns, A. Dennis, J. Speers, 2000. Rapid analysis of ecstasy and related phenethylamines in seized tablets by Raman spectroscopy. Analyst 125: 541-544.
  42. L .Taylor, F. Langkilde, 2000. Evaluation of solid-state forms present in tablets by Raman spectroscopy. J. Pharm. Sci. 89:1342-1353.
  43. A. Ringqvist, L .Taylor, K. Ekelund, G. Ragnarsson, S. Engström, A. Axelsson, 2003. Atomic force microscopy analysis and confocal Raman microimaging of coated pellets. Int. J. Pharm. 267: 35-47.
  44. H. Wikström, I. Lewis, L. Taylor, 2005. Comparison of sampling techniques for in-line monitoring using Raman spectroscopy. Submitted for publication.
  45. J. Johanson, S. Pettersson, L. Taylor, 2002. Infrared imaging of laser-induced heating during Raman spectroscopy of pharmaceutical solids. J. Pharm. Biomed. Anal. 20: 1223-1231.
  46. Raman, CV. The molecular scattering of light. Nobel Lecture, December 11, 1930.