Understanding Dissolution and Crystallization with Imaging: A Surface Point of ViewClick to copy article linkArticle link copied!
- Dunja Novakovic*Dunja Novakovic*E-mail: [email protected]Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014 Helsinki, FinlandMore by Dunja Novakovic
- Antti IsomäkiAntti IsomäkiBiomedicum Imaging Unit, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, FinlandMore by Antti Isomäki
- Bibi PleunisBibi PleunisDrug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014 Helsinki, FinlandMore by Bibi Pleunis
- Sara J. Fraser-MillerSara J. Fraser-MillerDodd-Walls Center for Photonic and Quantum Technologies, Department of Chemistry, University of Otago, Dunedin 9016, New ZealandMore by Sara J. Fraser-Miller
- Leena PeltonenLeena PeltonenDrug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014 Helsinki, FinlandMore by Leena Peltonen
- Timo LaaksonenTimo LaaksonenLaboratory of Chemistry and Bioengineering, Tampere University of Technology, Korkeakoulunkatu 8, 33720 Tampere, FinlandMore by Timo Laaksonen
- Clare J. StrachanClare J. StrachanDrug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014 Helsinki, FinlandMore by Clare J. Strachan
Abstract
The tendency for crystallization during storage and administration is the most considerable hurdle for poorly water-soluble drugs formulated in the amorphous form. There is a need to better detect often subtle and complex surface crystallization phenomena and understand their influence on the critical quality attribute of dissolution. In this study, the interplay between surface crystallization of the amorphous form during storage and dissolution testing, and its influence on dissolution behavior, is analyzed for the first time with multimodal nonlinear optical imaging (coherent anti-Stokes Raman scattering (CARS) and sum frequency generation (SFG)). Complementary analyses are provided with scanning electron microscopy, X-ray diffraction and infrared and Raman spectroscopies. Amorphous indomethacin tablets were prepared and subjected to two different storage conditions (30 °C/23% RH and 30 °C/75% RH) for various durations and then dissolution testing using a channel flow-through device. Trace levels of surface crystallinity previously imaged with nonlinear optics after 1 or 2 days of storage did not significantly decrease dissolution and supersaturation compared to the freshly prepared amorphous tablets while more extensive crystallization after longer storage times did. Multimodal nonlinear optical imaging of the tablet surfaces after 15 min of dissolution revealed complex crystallization behavior that was affected by both storage condition and time, with up to four crystalline polymorphs simultaneously observed. In addition to the well-known α- and γ-forms, the less reported metastable ε- and η-forms were also observed, with the ε-form being widely observed in samples that had retained significant surface amorphousness during storage. This form was also prepared in the pure form and further characterized. Overall, this study demonstrates the potential value of nonlinear optical imaging, together with more established solid-state analysis methods, to understand complex surface crystallization behavior and its influence on drug dissolution during the development of amorphous drugs and dosage forms.
1. Introduction
2. Materials and Methods
2.1. Sample Preparation
2.2. Intrinsic Dissolution Testing
2.3. Scanning Electron Microscopy (SEM)
2.4. X-ray Diffractometry (XRD)
2.5. Fourier Transform Infrared Spectroscopy (FTIR)
2.6. Multimodal Nonlinear Optical Imaging
3. Results
3.1. Dissolution Behavior
3.1.1. Pure Amorphous and Reference Crystalline Forms
Figure 1
Figure 1. Dissolution profiles (drug release over time) for samples stored at (a) 30 °C/23% RH and (b) 30 °C/75% RH. Profiles of the pure amorphous tablet (day 0) and reference crystalline γ- and α-forms are shown for comparison. Profiles are generated with mean values of a minimum of three measurements. Error bars represent plus one standard deviation.
3.1.2. Stored Samples
3.2. Visual Appearance
3.3. Surface Morphology
Figure 2
Figure 2. SEM micrographs of an amorphous indomethacin tablet (day 0, first column) and tablets stored for 1, 2, 7, and 22 days (from left to right) at 30 °C/23% RH before dissolution testing (top row). SEM images of the tablets prepared and stored in the same manner after 15 min of intrinsic dissolution testing (bottom row).
Figure 3
Figure 3. SEM micrographs of an amorphous indomethacin tablet (day 0, first column) and tablets stored for 1, 2, 7, and 22 days (from left to right) at 30 °C/75% RH before dissolution testing (top row). SEM images of the tablets prepared and stored in the same manner after 15 min of intrinsic dissolution testing (bottom row).
3.4. Solid-State Analysis
3.4.1. Freshly Prepared Amorphous Indomethacin (Day 0)
Figure 4
Figure 4. XRD diffractograms of tablets before and after 15 min of dissolution testing: (a) storage at 30 °C/23% RH and (b) storage at 30 °C/75% RH. Diffractograms of the α-, γ-, and ε-forms of indomethacin are shown for comparison, and some of their characteristic peaks are marked in green, red, and yellow, respectively. The ε-form was measured in transmission mode using a powder, while the α- and γ-forms were measured in reflection with tablets.
Figure 5
Figure 5. PCA scores plot of the ATR-FTIR spectra of the amorphous (day 0) and all stored samples after 15 min of dissolution testing (a); loadings and reference IR spectra of different solid-state forms of indomethacin (b).
Figure 6
Figure 6. CARS and SFG overlay images of samples stored at 30 °C/23% RH before (a) and after (b) 15 min of dissolution testing at pH 6.8. CARS spectra in the range of 1413–1800 cm–1 from selected regions marked by arrows plotted with reference spectra of amorphous, ε-, and γ-indomethacin (c–e). Overlay images (a, b) represent overlays of three channels: single CARS line at 1701 cm–1 in red (γ-indomethacin), single CARS line at 1676 cm–1 in blue (amorphous indomethacin), and a third channel representing the SFG signal (all noncentrosymmetric crystals) in green and yellow. The separation between green and yellow regions is based on the intensity ratios of CARS peaks at 1652 and 1676 cm–1 so that the regions having SFG activity and a CARS peak at 1652 cm–1 are colored green (α-indomethacin), and the regions having SFG activity and a CARS peak at 1676 cm–1 are colored yellow (ε-indomethacin). Panel (a) has been reprinted with permission from Novakovic et al. (18) Copyright 2017 American Chemical Society. am = amorphous.
Figure 7
Figure 7. CARS and SFG overlay images of samples stored at 30 °C/75% RH before (a) and after 15 min of dissolution testing at pH 6.8 (b). CARS spectra in the range of 1413–1800 cm–1 from selected regions marked by arrows (c–f) plotted with reference spectra of amorphous, ε-, α-, and γ-indomethacin. Overlay images (a, b) represent overlays of three channels: single CARS line at 1701 cm–1 in red (γ-indomethacin), single CARS line at 1676 cm–1 in blue (amorphous indomethacin), and a third channel representing the SFG signal (all noncentrosymmetric crystals) in green and yellow. The separation between green and yellow regions is based on the intensity ratios of CARS peaks at 1652 and 1676 cm–1 so that the regions having SFG activity and the CARS peak at 1652 cm–1 are colored green (α-indomethacin), and the regions having SFG activity and CARS peak at 1676 cm–1 are colored yellow (ε-indomethacin). Panel (a) has been reprinted with permission from Novakovic et al. (18) Copyright 2017 American Chemical Society. am = amorphous.
3.4.2. Tablets after Storage at Lower Humidity (30 °C/23% RH)
3.4.3. Tablets after Storage at Higher Humidity (30 °C/75% RH)
3.4.4. Reference Raman and CARS Spectra
Figure 8
Figure 8. CARS (solid lines) and Raman (dotted lines) spectra of indomethacin solid-state forms. Raman spectra (except for the ε-form) are reproduced with permission from ref (23). The CARS spectral resolution is 12 cm–1, while the Raman spectral resolution is 4 cm–1. All CARS spectra are measured from the prepared reference forms, except for the spectrum of the η-form, which was recorded at the tablet surface. Polymorphs marked with asterisk are SFG-active.
4. Discussion
sample | solid state forms | |||||
---|---|---|---|---|---|---|
XRD | ATR-FTIRa | CARS/SFG | ||||
before | after | before (18) | after | before (18) | after | |
Day 0 | amb | am, ε | am | am, ε (traces) | am | am, ε |
30 °C/23% RH | ||||||
Day 1 | am | am, ε | am | am, ε (traces) | am, γ | am, ε, γ |
Day 2 | am, γ | am, γ | am, γ | γ | am, γ, α (traces) | am, ε, γ |
Day 7 | γ | γ | γ | γ | γ, α | ε, γ |
Day 22 | γ | γ | γ | γ | γ, α (traces) | γ |
30 °C/75% RH | ||||||
Day 1 | am | am | am | am | am, α (traces) | am, ε, γ, α (traces) |
Day 2 | am, γ | am, γ, ε | am, γ | ε | am, α, γ (traces) | am, ε, γ, α |
Day 7 | am, γ, α | am, γ, α | am, γ | am, ε, γ | am, α, γ (traces) | am, ε, γ, α, η (traces) |
Day 22 | am, γ, α | am, γ, α | am, α | am, γ | am, α, γ (traces) | am, γ, α |
Tentative assignments based on IR spectra and PCA.
am = amorphous.
5. Conclusions
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.molpharmaceut.8b00840.
ATR-FTIR spectra of tablet surfaces after dissolution and additional CARS and SFG overlay images of tablets stored for 1 and 7 days at 30 °C/75% RH (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
Heikki Räikkönen from the University of Helsinki is gratefully acknowledged for his assistance with the XRD measurements. Marta Cristos from the The Complutense University of Madrid is acknowledged for her help in the initial intrinsic dissolution measurements and method setup. Jaana Koskela from the University of Helsinki is acknowledged for her help in preparing the δ-form of indomethacin. DN gratefully acknowledges the Doctoral Program in Drug Research funding provided by the University of Helsinki. CS acknowledges Business Finland, the Finnish Funding Agency for Innovation (project no. 1245/31/2015), the Academy of Finland (grant no. 289398), and the University of Helsinki for a three-year research project grant (2014–2016).
CARS | coherent anti-Stokes Raman scattering |
SFG | sum frequency generation |
SHG | second-harmonic generation |
SEM | scanning electron microscopy |
XRD | X-ray diffraction |
PLM | polarized light microscopy |
ATR-FTIR | attenuated total reflectance Fourier transform infrared spectroscopy |
DSC | differential scanning calorimetry |
References
This article references 47 other publications.
- 1European Directorate for the Quality of Medicines & HealthCare. European Pharmacopoeia 9.5. http://online6.edqm.eu/ep905/.Google ScholarThere is no corresponding record for this reference.
- 2Hancock, B. C.; Parks, M. What is the true solubility advantage for amorphous pharmaceuticals?. Pharm. Res. 2000, 17 (4), 397– 404, DOI: 10.1023/A:1007516718048Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXktFalsb0%253D&md5=995cc8852439cc0319b3112a6abbc021What is the true solubility advantage for amorphous pharmaceuticals?Hancock, Bruno C.; Parks, MichaelPharmaceutical Research (2000), 17 (4), 397-404CODEN: PHREEB; ISSN:0724-8741. (Kluwer Academic/Plenum Publishers)In order to evaluate the magnitude of the soly. advantage for amorphous pharmaceutical materials when compared to their cryst. counterparts, the thermal properties of several drugs in their amorphous and cryst. states were detd. using differential scanning calorimetry. From these properties the soly. advantage for the amorphous form was predicted as a function of temp. using a simple thermodn. anal. These predictions were compared to the results of exptl. measurements of the aq. solubilities of the amorphous and cryst. forms of the drugs at several temps. By treating each amorphous drug as either an equil. supercooled liq. or a pseudo-equil. glass, the soly. advantage compared to the most stable cryst. form was predicted to be between 10 and 1600 fold. The measured soly. advantage was usually considerably less than this, and for one compd. studied in detail its temp. dependence was also less than predicted. It was calcd. that even for partially amorphous materials the apparent soly. enhancement (theor. or measured) is likely to influence in-vitro and in-vivo dissoln. behavior. Amorphous pharmaceuticals are markedly more sol. than their cryst. counterparts, however, their exptl. soly. advantage is typically less than that predicted from simple thermodn. considerations. This appears to be the result of difficulties in detg. the soly. of amorphous materials under true equil. conditions. Simple thermodn. predictions can provide a useful indication of the theor. max. soly. advantage for amorphous pharmaceuticals, which directly reflects the driving force for their initial dissoln.
- 3Murdande, S. B.; Pikal, M. J.; Shanker, R. M.; Bogner, R. H. Solubility advantage of amorphous pharmaceuticals: I. A thermodynamic analysis. J. Pharm. Sci. 2010, 99 (3), 1254– 1264, DOI: 10.1002/jps.21903Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmvVCqtg%253D%253D&md5=215a9f5a55ea378793b3eec9c263590aSolubility advantage of amorphous pharmaceuticals: I. A thermodynamic analysisMurdande, Sharad B.; Pikal, Michael J.; Shanker, Ravi M.; Bogner, Robin H.Journal of Pharmaceutical Sciences (2010), 99 (3), 1254-1264CODEN: JPMSAE; ISSN:0022-3549. (Wiley-Liss, Inc.)In recent years there has been growing interest in advancing amorphous pharmaceuticals as an approach for achieving adequate soly. Due to difficulties in the exptl. measurement of soly., a reliable est. of the soly. enhancement ratio of an amorphous form of a drug relative to its cryst. counterpart would be highly useful. We have developed a rigorous thermodn. approach to est. enhancement in soly. that can be achieved by conversion of a cryst. form to the amorphous form. We rigorously treat the three factors that contribute to differences in soly. between amorphous and cryst. forms. First, we calc. the free energy difference between amorphous and cryst. forms from thermal properties measured by modulated differential scanning calorimetry (MDSC). Secondly, since an amorphous solute can absorb significant amts. of water, which reduces its activity and soly., a correction is made using water sorption isotherm data and the Gibbs-Duhem equation. Next, a correction is made for differences in the degree of ionization due to differences in solubilities of the two forms. Utilizing this approach the theor. estd. soly. enhancement ratio of 7.0 for indomethacin (amorphous/γ-crystal) was found to be in close agreement with the exptl. detd. ratio of 4.9. © 2009 Wiley-Liss, Inc. and the American Pharmacists Assocn. J Pharm Sci 99: 1254-1264, 2010.
- 4Debnath, S.; Predecki, P.; Suryanarayanan, R. Use of glancing angle x-ray powder diffractometry to depth-profile phase transformations during dissolution of indomethacin and theophylline tablets. Pharm. Res. 2004, 21 (1), 149– 159, DOI: 10.1023/B:PHAM.0000012163.89163.f8Google ScholarThere is no corresponding record for this reference.
- 5Ewing, A. V.; Clarke, G. S.; Kazarian, S. G. Stability of indomethacin with relevance to the release from amorphous solid dispersions studied with ATR-FTIR spectroscopic imaging. Eur. J. Pharm. Sci. 2014, 60, 64– 71, DOI: 10.1016/j.ejps.2014.05.001Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXpvVagtbc%253D&md5=f0c5aea53b60d21b42d8da8076eebb6eStability of indomethacin with relevance to the release from amorphous solid dispersions studied with ATR-FTIR spectroscopic imagingEwing, Andrew V.; Clarke, Graham S.; Kazarian, Sergei G.European Journal of Pharmaceutical Sciences (2014), 60 (), 64-71CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)This work presents the use of attenuated total reflection Fourier transform IR (ATR-FTIR) spectroscopy and spectroscopic imaging to study the stability and dissoln. behavior of amorphous solid dispersions (ASDs). ASDs are employed to improve the bioavailability of drugs which are poorly sol. in aq. solns. Selecting the appropriate polymeric excipients for use in pharmaceutical tablets is crucial to control drug stability and subsequent release. In this study, indomethacin was used as a model poorly-aq. sol. drug since the amorphous-form has improved dissoln. properties over its cryst. forms. ASDs of indomethacin/polyethylene glycol (PEG) and indomethacin/hydroxypropyl methylcellulose (HPMC) in a 1:3 wt ratio were compared. Firstly, ATR-FTIR spectroscopy was employed to monitor the stability of indomethacin in the ASDs over 96 h. While the indomethacin/HPMC ASD showed the ability to maintain the amorphous indomethacin form for longer periods of time, ATR-FTIR spectra revealed that indomethacin in the drug/PEG ASD crystd. to the stable γ-form, via the α-form. Secondly, ATR-FTIR spectroscopic imaging was used to study the dissoln. of ASD tablets in a phosphate buffer (pH 7.5). Crystn. of amorphous indomethacin was characterized in the spectra collected during the dissoln. of the indomethacin/PEG ASD which consequently hindered release into the surrounding soln. In contrast, release of amorphous indomethacin was more effective from HPMC.
- 6Savolainen, M.; Kogermann, K.; Heinz, A.; Aaltonen, J.; Peltonen, L.; Strachan, C.; Yliruusi, J. Better understanding of dissolution behaviour of amorphous drugs by in situ solid-state analysis using Raman spectroscopy. Eur. J. Pharm. Biopharm. 2009, 71 (1), 71– 79, DOI: 10.1016/j.ejpb.2008.06.001Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsV2lurfI&md5=4cbb70cb1088b3c72be715a8e848f0edBetter understanding of dissolution behaviour of amorphous drugs by in situ solid-state analysis using Raman spectroscopySavolainen, M.; Kogermann, K.; Heinz, A.; Aaltonen, J.; Peltonen, L.; Strachan, C.; Yliruusi, J.European Journal of Pharmaceutics and Biopharmaceutics (2009), 71 (1), 71-79CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)Amorphous drugs have a higher kinetic soly. and dissoln. rate than their cryst. counterparts. However, this advantage is lost if the amorphous form converts to the stable cryst. form during the dissoln. as the dissoln. rate will gradually change to that of the cryst. form. The purpose of this study was to use in situ Raman spectroscopy in combination with either partial least squares discriminant anal. (PLS-DA) or partial least squares (PLS) regression anal. to monitor as well as quantify the solid-phase transitions that take place during the dissoln. of two amorphous drugs, indomethacin (IMC) and carbamazepine (CBZ). The dissoln. rate was higher from amorphous IMC compared to the cryst. α- and γ-forms. However, the dissoln. rate started to slow down during the expt. In situ Raman anal. verified that at that time point the sample started to crystallize to the α-form. Amorphous CBZ instantly started to crystallize upon contact with the dissoln. medium. The transition from the amorphous form to CBZ dihydrate appears to go through the anhydrate form I. Based on the PLS anal. the amt. of form I formed in the sample during the dissoln. affected the dissoln. rate. Raman spectroscopy combined with PLS-DA was also more sensitive to the solid-state changes than X-ray powder diffraction (XRPD) and was able to detect changes in the solid-state that could not be detected with XRPD.
- 7Aaltonen, J.; Heinänen, P.; Peltonen, L.; Kortejärvi, H.; Tanninen, V. P.; Christiansen, L.; Hirvonen, J.; Yliruusi, J.; Rantanen, J. In situ measurement of solvent-mediated phase transformations during dissolution testing. J. Pharm. Sci. 2006, 95 (12), 2730– 2737, DOI: 10.1002/jps.20725Google ScholarThere is no corresponding record for this reference.
- 8Greco, K.; Bogner, R. Crystallization of amorphous indomethacin during dissolution: Effect of processing and annealing. Mol. Pharmaceutics 2010, 7 (5), 1406– 1418, DOI: 10.1021/mp1000197Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmt1Kruro%253D&md5=4047d8e6882e5d4f5d9f001ce1f2c948Crystallization of Amorphous Indomethacin during Dissolution: Effect of Processing and AnnealingGreco, Kristyn; Bogner, RobinMolecular Pharmaceutics (2010), 7 (5), 1406-1418CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The crystn. of amorphous drugs during dissoln. is a type of soln. mediated phase transformation that can reduce the bioavailability enhancement one hoped to gain from the amorphous state. The goal of this study was to explore the effects of processing on the dissoln. performance of amorphous indomethacin. The amorphous solids were prepd. by four techniques, quench cooling the melted solid, cryogrinding γ indomethacin amorphous for 1 or 3 h and quench cooling the solid followed by 1 h of cryogrinding. Dissoln. results assessed in a flow-through intrinsic dissoln. app. reveal decreases in the dissoln. rate of amorphous indomethacin during the exptl. time frame indicating that a soln. mediated phase transformation has occurred. The amorphous solids prepd. by melt quenching and melt quenching followed by cryogrinding showed a significant dissoln. rate advantage over the γ form of indomethacin. In contrast, indomethacin that was cryoground amorphous for 1 or 3 h did not show any dissoln. rate advantage over the cryst. material. Transformation was confirmed by in situ Raman microscopy and polarized light microscopy with differences seen in the nature of the crystals apparent on the surface of the dissolving solid. A portion of the melt quenched amorphous sample was annealed at 25°C and 0% relative humidity to induce partial crystn. of γ indomethacin. As crystallinity increased, the dissoln. rate decreased. The transformation time of partially amorphous indomethacin was not dependent on the level of crystallinity present, indicating only a small fraction of cryst. material needs to be present to affect the kinetics of crystn. The soln. mediated phase transformation of amorphous indomethacin is affected by the processing method even though all solids were confirmed amorphous by polarized light microscopy and X-ray diffraction. Dissoln. may distinguish differences in amorphous solids that other methods cannot discern.
- 9Schmitt, P. D. Recent advances in nonlinear optical analyses of pharmaceutical materials in the solid state. Mol. Pharmaceutics 2017, 14 (3), 555– 565, DOI: 10.1021/acs.molpharmaceut.6b00809Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1Srsb8%253D&md5=55995e7bda16050a15aba0d39bc054c0Recent Advances in Nonlinear Optical Analyses of Pharmaceutical Materials in the Solid StateSchmitt, Paul D.Molecular Pharmaceutics (2017), 14 (3), 555-565CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)A review. The past decade has seen an increase in the use of nonlinear optical (NLO) techniques such as second harmonic generation, coherent antistokes Raman scattering, stimulated Raman scattering, and two-photon fluorescence for the solid-state characterization of pharmaceutical materials. These combined techniques offer several advantages (e.g., speed, selectivity, quantitation) of potential interest to the pharmaceutical community, as decreased characterization times in formulation development and testing could help decrease the time required to bring new, higher quality drugs to market. The large body of literature recently published in this field merits a review. Literature will be discussed in order of drug development, starting with applications in initial therapeutic mol. crystn. and polymorphic anal., followed by final dosage form characterization, and ending with drug product performance testing.
- 10Windbergs, M.; Jurna, M.; Offerhaus, H. L.; Herek, J. L.; Kleinebudde, P.; Strachan, C. J. Chemical imaging of oral solid dosage forms and changes upon dissolution using coherent anti-Stokes Raman scattering microscopy. Anal. Chem. 2009, 81 (6), 2085– 2091, DOI: 10.1021/ac8020856Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhslCisrc%253D&md5=4ecd9d725f53618dabc5d25210fc988aChemical imaging of oral solid dosage forms and changes upon dissolution using coherent anti-Stokes Raman scattering microscopyWindbergs, Maike; Jurna, Martin; Offerhaus, Herman L.; Herek, Jennifer L.; Kleinebudde, Peter; Strachan, Clare J.Analytical Chemistry (Washington, DC, United States) (2009), 81 (6), 2085-2091CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Dissoln. testing is a crucial part of pharmaceutical dosage form investigations and is generally performed by analyzing the concn. of the released drug in a defined vol. of flowing dissoln. medium. As solid-state properties of the components affect dissoln. behavior to a large and sometimes even unpredictable extent there is a strong need for monitoring and esp. visualizing solid-state properties during dissoln. testing. In this study coherent anti-Stokes Raman scattering (CARS) microscopy was used to visualize the solid-state properties of lipid-based oral dosage forms contg. the model drug theophylline anhydrate during dissoln. in real time. The drug release from the dosage form matrix was monitored with a spatial resoln. of about 1.5 μm. In addn., as theophylline anhydrate tends to form the less sol. monohydrate during dissoln., CARS microscopy allowed the solid-state transformation of the drug to be spatially visualized. The results obtained by CARS microscopy revealed that the method used to combine lipid and active ingredient into a sustained release dosage form can influence the physicochem. behavior of the drug during dissoln. In this case, formation of theophylline monohydrate on the surface was visualized during dissoln. with tablets compressed from powd. mixts. but not with solid lipid extrudates.
- 11Jurna, M.; Windbergs, M.; Strachan, C. J.; Hartsuiker, L.; Otto, C.; Kleinebudde, P.; Herek, J. L.; Offerhaus, H. L. Coherent anti-Stokes Raman scattering microscopy to monitor drug dissolution in different oral pharmaceutical tablets. J. Innovative Opt. Health Sci. 2009, 2 (1), 37– 43, DOI: 10.1142/S1793545809000322Google ScholarThere is no corresponding record for this reference.
- 12Fussell, A.; Garbacik, E.; Offerhaus, H.; Kleinebudde, P.; Strachan, C. In situ dissolution analysis using coherent anti-Stokes Raman scattering (CARS) and hyperspectral CARS microscopy. Eur. J. Pharm. Biopharm. 2013, 85 (3, Part B), 1141– 1147, DOI: 10.1016/j.ejpb.2013.08.012Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVylurrF&md5=e2c5dba6649cdfb115c1fd8c43765d3eIn situ dissolution analysis using coherent anti-Stokes Raman scattering (CARS) and hyperspectral CARS microscopyFussell, Andrew; Garbacik, Erik; Offerhaus, Herman; Kleinebudde, Peter; Strachan, ClareEuropean Journal of Pharmaceutics and Biopharmaceutics (2013), 85 (3PB), 1141-1147CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)The solid-state form of an active pharmaceutical ingredient (API) in an oral dosage form plays an important role in detg. the dissoln. rate of the API. As the solid-state form can change during dissoln., there is a need to monitor the oral dosage form during dissoln. testing. Coherent anti-Stokes Raman scattering (CARS) microscopy provides rapid, spectrally selective imaging to monitor the oral dosage form during dissoln. In this study, in situ CARS microscopy was combined with inline UV absorption spectroscopy to monitor the solid-state change in oral dosage forms contg. theophylline anhydrate undergoing dissoln. and to correlate the solid-state change with a change in dissoln. rate. The results from in situ CARS microscopy showed that theophylline anhydrate converted to theophylline monohydrate during dissoln. resulting in a redn. in the dissoln. rate. The addn. of Me cellulose to the dissoln. medium was found to delay the theophylline monohydrate growth and changed the morphol. of the monohydrate. The net effect was an increased dissoln. rate for theophylline anhydrate. Our results show that in situ CARS microscopy combined with inline UV absorption spectroscopy is capable of monitoring oral dosage forms undergoing dissoln. and correlating changes in solid-state form with changes in dissoln. rate.
- 13Kestur, U. S.; Wanapun, D.; Toth, S. J.; Wegiel, L. A.; Simpson, G. J.; Taylor, L. S. Nonlinear optical imaging for sensitive detection of crystals in bulk amorphous powders. J. Pharm. Sci. 2012, 101 (11), 4201– 4213, DOI: 10.1002/jps.23280Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFWmtrnN&md5=8786157ff38e6777dea6847f3adf68feNonlinear optical imaging for sensitive detection of crystals in bulk amorphous powdersKestur, Umesh S.; Wanapun, Duangporn; Toth, Scott J.; Wegiel, Lindsay A.; Simpson, Garth J.; Taylor, Lynne S.Journal of Pharmaceutical Sciences (2012), 101 (11), 4201-4213CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)The primary aim of this study was to evaluate the utility of second-order nonlinear imaging of chiral crystals (SONICC) to quantify crystallinity in drug-polymer blends, including solid dispersions. Second harmonic generation (SHG) can potentially exhibit scaling with crystallinity between linear and quadratic depending on the nature of the source, and thus, it is important to det. the response of pharmaceutical powders. Phys. mixts. contg. different proportions of cryst. naproxen and hydroxyl Pr Me cellulose acetate succinate (HPMCAS) were prepd. by blending and a dispersion was produced by solvent evapn. A custom-built SONICC instrument was used to characterize the SHG intensity as a function of the cryst. drug fraction in the various samples. Powder X-ray diffraction (PXRD) and Raman spectroscopy were used as complementary methods known to exhibit linear scaling. SONICC was able to detect cryst. drug even in the presence of 99.9 wt % HPMCAS in the binary mixts. The calibration curve revealed a linear dynamic range with a R2 value of 0.99 spanning the range from 0.1 to 100 wt % naproxen with a root mean square error of prediction of 2.7%. Using the calibration curve, the errors in the validation samples were in the range of 5%-10%. Anal. of a 75 wt % HPMCAS-naproxen solid dispersion with SONICC revealed the presence of crystallites at an earlier time point than could be detected with PXRD and Raman spectroscopy. In addn., results from the crystn. kinetics expt. using SONICC were in good agreement with Raman spectroscopy and PXRD. In conclusion, SONICC has been found to be a sensitive technique for detecting low levels (0.1% or lower) of crystallinity, even in the presence of large quantities of a polymer. © 2012 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 14Wanapun, D.; Kestur, U. S.; Kissick, D. J.; Simpson, G. J.; Taylor, L. S. Selective detection and quantitation of organic molecule crystallization by second harmonic generation microscopy. Anal. Chem. 2010, 82 (13), 5425– 5432, DOI: 10.1021/ac100564fGoogle Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmvVSmsLo%253D&md5=1d176cb9495fe9eff0336a0faf80bd57Selective Detection and Quantitation of Organic Molecule Crystallization by Second Harmonic Generation MicroscopyWanapun, Duangporn; Kestur, Umesh S.; Kissick, David J.; Simpson, Garth J.; Taylor, Lynne S.Analytical Chemistry (Washington, DC, United States) (2010), 82 (13), 5425-5432CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Second order nonlinear optical imaging of chiral crystals (SONICC) was applied to selectively detect crystal formation at early stages and characterize the kinetics of nucleation and growth. SONICC relies on second harmonic generation (SHG), a nonlinear optical effect that only arises from noncentosym. ordered domain structures, which include crystals of chiral mols. The model systems studied include pharmaceutically relevant compds.: griseofulvin and chlorpropamide. SONICC demonstrates low detection limits producing an 8 order of magnitude improvement relative to macroscopic av. techniques and 5 order of magnitude improvement relative to optical microscopy. SONICC was also applied to examine the kinetics of crystn. in amorphous griseofulvin. The results show that SONICC enables simultaneous monitoring of individual crystal growth, nucleation rate, and macroscopic crystn. kinetics.
- 15Schmitt, P. D.; Trasi, N. S.; Taylor, L. S.; Simpson, G. J. Finding the needle in the haystack: Characterization of trace crystallinity in a commercial formulation of paclitaxel protein-bound particles by Raman spectroscopy enabled by second harmonic generation microscopy. Mol. Pharmaceutics 2015, 12 (7), 2378– 2383, DOI: 10.1021/acs.molpharmaceut.5b00065Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVSht7nM&md5=244ef3d7eb97c0f6fdae03db367bb73dFinding the Needle in the Haystack: Characterization of Trace Crystallinity in a Commercial Formulation of Paclitaxel Protein-Bound Particles by Raman Spectroscopy Enabled by Second Harmonic Generation MicroscopySchmitt, Paul D.; Trasi, Niraj S.; Taylor, Lynne S.; Simpson, Garth J.Molecular Pharmaceutics (2015), 12 (7), 2378-2383CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Second harmonic generation (SHG) microscopy was used to rapidly identify regions of interest for localized confocal Raman spectroscopy measurements in order to quantify crystallinity within lyophilized Abraxane powder (protein bound paclitaxel for injectable suspension). Water insol. noncentrosym. cryst. particles ranging from ∼1 to 120 μm were identified by SHG, with wide variability in crystal size and frequency obsd. between several batches of Abraxane. By targeting the Raman anal. to these localized regions identified by SHG, the required measurement time was decreased over 2 orders of magnitude, from 8 h to 2 s. Exptl. Raman spectra of SHG active domains in Abraxane were in good agreement with exptl. spectra of pure cryst. paclitaxel. These collective results are consistent with up to 30% of the active ingredient being present as poorly sol. cryst. particulates in some batches of Abraxane.
- 16Mah, P. T.; Novakovic, D.; Saarinen, J.; Van Landeghem, S.; Peltonen, L.; Laaksonen, T.; Isomäki, A.; Strachan, C. J. Elucidation of compression-induced surface crystallization in amorphous tablets using sum frequency generation (SFG) microscopy. Pharm. Res. 2017, 34 (5), 957– 970, DOI: 10.1007/s11095-016-2046-6Google ScholarThere is no corresponding record for this reference.
- 17Song, Z.; Sarkar, S.; Vogt, A. D.; Danzer, G. D.; Smith, C. J.; Gualtieri, E. J.; Simpson, G. J. Kinetic modeling of accelerated stability testing enabled by second harmonic generation microscopy. Anal. Chem. 2018, 90 (7), 4406– 4413, DOI: 10.1021/acs.analchem.7b04260Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvFGns7w%253D&md5=a534778ce5b78e77924df5ba76416292Kinetic Modeling of Accelerated Stability Testing Enabled by Second Harmonic Generation MicroscopySong, Zhengtian; Sarkar, Sreya; Vogt, Andrew D.; Danzer, Gerald D.; Smith, Casey J.; Gualtieri, Ellen J.; Simpson, Garth J.Analytical Chemistry (Washington, DC, United States) (2018), 90 (7), 4406-4413CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)The low limits of detection afforded by second harmonic generation (SHG) microscopy coupled with image anal. algorithms enabled quant. modeling of the temp.-dependent crystn. of active pharmaceutical ingredients (APIs) within amorphous solid dispersions (ASDs). ASDs, in which an API is maintained in an amorphous state within a polymer matrix, are finding increasing use to address soly. limitations of small-mol. APIs. Extensive stability testing is typically performed for ASD characterization, the time frame for which is often dictated by the earliest detectable onset of crystal formation. Here a study of accelerated stability testing on ritonavir, a human immunodeficiency virus (HIV) protease inhibitor, has been conducted. Under the condition for accelerated stability testing at 50 °C/75%RH and 40 °C/75%RH, ritonavir crystn. kinetics from amorphous solid dispersions were monitored by SHG microscopy. SHG microscopy coupled by image anal. yielded limits of detection for ritonavir crystals as low as 10 ppm, which is about 2 orders of magnitude lower than other methods currently available for crystallinity detection in ASDs. The four decade dynamic range of SHG microscopy enabled quant. modeling with an established (JMAK) kinetic model. From the SHG images, nucleation and crystal growth rates were independently detd.
- 18Novakovic, D.; Saarinen, J.; Rojalin, T.; Antikainen, O.; Fraser-Miller, S. J.; Laaksonen, T.; Peltonen, L.; Isomäki, A.; Strachan, C. J. Multimodal nonlinear optical imaging for sensitive detection of multiple pharmaceutical solid-state forms and surface transformations. Anal. Chem. 2017, 89 (21), 11460– 11467, DOI: 10.1021/acs.analchem.7b02639Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFOjt7rE&md5=02aa9d1d14293c87a2ac665153bfdde8Multimodal Nonlinear Optical Imaging for Sensitive Detection of Multiple Pharmaceutical Solid-State Forms and Surface TransformationsNovakovic, Dunja; Saarinen, Jukka; Rojalin, Tatu; Antikainen, Osmo; Fraser-Miller, Sara J.; Laaksonen, Timo; Peltonen, Leena; Isomaki, Antti; Strachan, Clare J.Analytical Chemistry (Washington, DC, United States) (2017), 89 (21), 11460-11467CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Two nonlinear imaging modalities, coherent anti-Stokes Raman scattering (CARS) and sum-frequency generation (SFG), were successfully combined for sensitive multimodal imaging of multiple solid-state forms and their changes on drug tablet surfaces. Two imaging approaches were used and compared: (i) hyperspectral CARS combined with principal component anal. (PCA) and SFG imaging and (ii) simultaneous narrowband CARS and SFG imaging. Three different solid-state forms of indomethacin-the cryst. gamma and alpha forms, as well as the amorphous form-were clearly distinguished using both approaches. Simultaneous narrowband CARS and SFG imaging was faster, but hyperspectral CARS and SFG imaging has the potential to be applied to a wider variety of more complex samples. These methodologies were further used to follow crystn. of indomethacin on tablet surfaces under two storage conditions: 30 °C/23% RH and 30 °C/75% RH. Imaging with (sub)micron resoln. showed that the approach allowed detection of very early stage surface crystn. The surfaces progressively crystd. to predominantly (but not exclusively) the gamma form at lower humidity and the alpha form at higher humidity. Overall, this study suggests that multimodal nonlinear imaging is a highly sensitive, solid-state (and chem.) specific, rapid, and versatile imaging technique for understanding and hence controlling (surface) solid-state forms and their complex changes in pharmaceuticals.
- 19Yamamoto, H. 1-Acyl-indoles. II. A new syntheses of 1-(p-chlorobenzoyl)-5-methoxy-3-indolylacetic acid and its polymorphism. Chem. Pharm. Bull. 1968, 16 (1), 17– 19, DOI: 10.1248/cpb.16.17Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaF1cXkt1OitL0%253D&md5=3b9da89d4aaa5f1bd18f35caa43da1a21-Acyl-indoles. II. A new syntheses of 1-(ion-chlorobenzoyl)-5-methoxy-3-indolylacetic acid and its polymorphismYamamoto, HisaoChemical & Pharmaceutical Bulletin (1968), 16 (1), 17-19CODEN: CPBTAL; ISSN:0009-2363.1-(p-Chlorobenzoyl)-2-methyl-5-methoxy-3-indolylacetic acid, a potent antiinflammatory drug, was directly prepd. from N-(p-chlorobenzoyl)-p-methoxyphenyl-hydrazine-HCl and levulinic acid in excellent yield by the new method. The recrystn. of 1-(p-chlorobenzoyl)-2-methyl-5-methoxy-3-indolylacetic acid from solvents gave crystals of polymorphism -α, β, and γ-types.
- 20Borka, L. The Polymorphism of indomethacin. New modifications, their melting behavior and solubility. Acta Pharm. Suec. 1974, 11 (3), 295– 303Google ScholarThere is no corresponding record for this reference.
- 21Lin, S. Y. Isolation and solid-state characteristics of a new crystal form of indomethacin. J. Pharm. Sci. 1992, 81 (6), 572– 576, DOI: 10.1002/jps.2600810622Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XkvFOiu7c%253D&md5=efd5a63b7b98cc610591f884fb4c662aIsolation and solid-state characteristics of a new crystal form of indomethacinLin, Shan YangJournal of Pharmaceutical Sciences (1992), 81 (6), 572-6CODEN: JPMSAE; ISSN:0022-3549.A new polymorphic crystal form of indomethacin was pptd. from an aq. soln. of indomethacin and β-cyclodextrin by titrn. with a 0.5N HCl aq. soln. Three polymorphs (α, β, and γ forms) and a new crystal form were differentiated with thermal anal., IR spectroscopy, powder x-ray diffractometry, TLC, elemental anal., and Fourier-transform IR (FTIR) spectroscopy with a newly developed FTIR microscope equipped with a thermal analyzer. The new crystal polymorph of indomethacin exhibited endo- and exothermic peaks near 102.8 and 104.1°, resp., because of phase transition without wt. loss, followed by 2 addnl. endothermic peaks at 151 and 158.9°, because of fusion. The differential scanning calorimetry-FTIR system can be used to examine the correlation between structural change of C:O stretching bands of this new polymorph and its thermal response. The new crystal form contained some γ-form crystals, detd. with an FTIR microscope equipped with a mapping option. Solid-state 13C-NMR spectra of the polymorphs of indomethacin were also examd.
- 22Crowley, K. J.; Zografi, G. Cryogenic grinding of indomethacin polymorphs and solvates: Assessment of amorphous phase formation and amorphous phase physical stability. J. Pharm. Sci. 2002, 91 (2), 492– 507, DOI: 10.1002/jps.10028Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XhsVSms7g%253D&md5=c8cde5407ebb09c5062ae2debff18c39Cryogenic grinding of indomethacin polymorphs and solvates: assessment of amorphous phase formation and amorphous phase physical stabilityCrowley, Kieran J.; Zografi, GeorgeJournal of Pharmaceutical Sciences (2002), 91 (2), 492-507CODEN: JPMSAE; ISSN:0022-3549. (Wiley-Liss, Inc.)The effect of cryogenic grinding on five crystal forms of indomethacin (IMC) was investigated with particular interest in the formation of amorphous phase. Powder x-ray diffraction (PXRD) and differential scanning calorimetry (DSC) demonstrated that amorphous phase formation took place for all three polymorphs (γ, α, and δ) and one solvate (IMC methanolate). In the latter case, a postgrinding drying stage was needed to remove desolvated methanol from the ground amorphous product because methanol destabilized amorphous IMC presumably via a plasticizing effect. The crystal structure of another solvate, IMC t-butanolate, was unaffected by grinding, indicating that amorphous phase formation on grinding does not occur in all cases. Ground amorphous materials possessed similar glass transition temps. but significant differences in phys. stability as assessed by both isothermal and nonisothermal crystn. It is argued that phys. factors, namely residual crystal phase and sp. surface area, det. the isothermal and nonisothermal crystn. behavior of ground amorphous samples as opposed to intrinsic differences in the structure of the amorphous phase.
- 23Surwase, S. A.; Boetker, J. P.; Saville, D.; Boyd, B. J.; Gordon, K. C.; Peltonen, L.; Strachan, C. J. Indomethacin: New polymorphs of an old drug. Mol. Pharmaceutics 2013, 10 (12), 4472– 4480, DOI: 10.1021/mp400299aGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVWkur%252FI&md5=1d1974f7636c482223c74202c21af783Indomethacin: New Polymorphs of an Old DrugSurwase, Sachin A.; Boetker, Johan P.; Saville, Dorothy; Boyd, Ben J.; Gordon, Keith C.; Peltonen, Leena; Strachan, Clare J.Molecular Pharmaceutics (2013), 10 (12), 4472-4480CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)This study reports the appearance and characterization of multiple new polymorphic forms of indomethacin. Considering the interest in amorphous suspensions for toxicol. studies of poorly water-sol. drugs, we sought to investigate the crystn. behavior of amorphous indomethacin in aq. suspension. Specifically, the effect of pH and temp. on crystn. behavior was studied. Quench cooled amorphous powder was added to buffered media at different pH values (1.2, 4.5, and 6.8) at 5 and 25 °C. Both the solid and the soln. were analyzed at different time points up to 24 h. Attenuated total reflection Fourier transform IR (ATR-FTIR) spectroscopy (with principal component anal.) was used to study solid-phase transformations and UV spectroscopy used to probe soln. concn. The crystn. onset time decreased and rate of crystn. increased with increasing pH and temp. Diverse polymorphic forms were obsd., with three new forms being identified; these were named ε, ζ, and η. At 25 °C, the amorphous form recrystd. directly to the α form, except at pH 6.8, where it initially converted briefly into the ε form. At 5 °C, all three new polymorphic forms were obsd. sequentially in the order ε, ζ, and then η, with the no. of these forms obsd. increasing sequentially with decreasing pH. The three new forms exhibited distinct X-ray powder diffraction (XPRD), differential scanning calorimetry (DSC), and FTIR and Raman spectroscopy profiles. The soln. concn. profiles suggest that the relative phys. stabilities of the polymorphs at 5 °C from lowest to highest is ε < ζ < η < α. The appearance of new polymorphs in this study suggests that amorphous suspensions are worth considering when performing polymorphic screening studies.
- 24Van Duong, T.; Ludeker, D.; Van Bockstal, P. J.; De Beer, T.; Van Humbeeck, J.; Van den Mooter, G. Polymorphism of indomethacin in semicrystalline dispersions: Formation, transformation, and segregation. Mol. Pharmaceutics 2018, 15 (3), 1037– 1051, DOI: 10.1021/acs.molpharmaceut.7b00930Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjs1agtQ%253D%253D&md5=3c27a34d72f18434d3e46ab6f38c4c0dPolymorphism of Indomethacin in Semicrystalline Dispersions: Formation, Transformation, and SegregationVan Duong, Tu; Ludeker, David; Van Bockstal, Pieter-Jan; De Beer, Thomas; Van Humbeeck, Jan; Van den Mooter, GuyMolecular Pharmaceutics (2018), 15 (3), 1037-1051CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The crystn. of metastable crystal polymorphs in polymer matrixes has been extensively reported in literature as a possible approach to enhance the soly. of poorly water-sol. drug compds., yet no clarification of the mechanism of the polymorph formation has been proposed. The current work aims to elucidate the polymorphism behavior of the model compd. indomethacin as well as the mechanism of polymorph selection of drugs in semicryst. systems. Indomethacin crystd. as either the α- or τ-form, a new metastable form, or a mixt. of the two polymorphs in dispersions contg. different drug loadings in polyethylene glycol, poloxamer, or Gelucire as the result of the variation in the mobility of drug mols. As a general rule, low mol. mobility of the amorphous drug favors the crystn. into thermodynamically stable forms whereas metastable cryst. polymorphs are preferred when the mol. mobility of the drug is sufficiently high. This rule provides insight into the polymorph selection of numerous active pharmaceutical ingredients in semicryst. dispersions and can be used as a guide for polymorphic screening from melt crystn. by tuning the mobility of drug mols. In addn., the drug crystd. faster while the polymer crystd. slower as the drug-loading increased with the maxima of drug crystn. rate in 70% indomethacin dispersion. Increasing the drug content in solid dispersions reduced the τ to α polymorphic transition rate, except for when the more stable form was initially dominant. The segregation of τ and α polymorphs as well as the polymorphic transformation during storage led to the inherent inhomogeneity of the semicryst. dispersions. This study highlights and expands our understanding about the complex crystn. behavior of semicryst. systems and is crucial for prepn. of solid dispersions with reproducible and consistent physicochem. properties and pharmaceutical performance.
- 25Kaneniwa, N.; Otsuka, M.; Hayashi, T. Physicochemical characterization of indomethacin polymorphs and the transformation kinetics in ethanol. Chem. Pharm. Bull. 1985, 33 (8), 3447– 3455, DOI: 10.1248/cpb.33.3447Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2MXmtVSrsrw%253D&md5=51415a5ce87b23c8d87e361fc1052889Physicochemical characterization of indomethacin polymorphs and the transformation kinetics in ethanolKaneniwa, Nobuyoshi; Otsuka, Makoto; Hayashi, TetsuoChemical & Pharmaceutical Bulletin (1985), 33 (8), 3447-55CODEN: CPBTAL; ISSN:0009-2363.Methods for the prepn. of polymorphs of indomethacin (IMC) (I) [53-86-1] were studied to obtain the pure polymorphs. The physicochem. properties of IMC polymorphs were measured by using x-ray diffraction anal. IR spectroscopy, DTA DSC, and 2 polymorphs (α and γ forms) and 1 benzene solvate (β form) were identified. The pure α form was obtained when distd. water at room temp. was poured into an EtOH [64-17-5] soln. of IMC at ∼80°, and the pptd. crystals were filtered and dried. The pure β and γ forms were obtained by recrystn. from benzene and Et2O, resp., at room temp. The m.ps. of the α and γ forms were 148 and 154°, resp., and their heats of fusion were 7.49 and 8.64 kcal/mol, resp., as detd. by DSC. A mixt. of α and γ forms was obtained by the method previously reported for α form prepn. (recrystn. method), since the pure α form was transformed to the γ form in EtOH at room temp. The transformation of α form to γ form in EtOH was analyzed by the kinetic method using 9 kinds of kinetic models. The transformation followed kinetics corresponding to 2-dimensional growth of nuclei (Avrami equation), and the activation energy was 14.2 kcal/mol from the Arrhenius plot. The solubilities of the α and γ forms in distd. water were 0.87 and 0.69 mg/100 mL, resp.
- 26Karmwar, P.; Graeser, K.; Gordon, K. C.; Strachan, C. J.; Rades, T. Effect of different preparation methods on the dissolution behaviour of amorphous indomethacin. Eur. J. Pharm. Biopharm. 2012, 80 (2), 459– 464, DOI: 10.1016/j.ejpb.2011.10.006Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XitVCisLk%253D&md5=5dd34ccc6eeaea3d7228f6eb742d3ca4Effect of different preparation methods on the dissolution behaviour of amorphous indomethacinKarmwar, Pranav; Graeser, Kirsten; Gordon, Keith C.; Strachan, Clare J.; Rades, ThomasEuropean Journal of Pharmaceutics and Biopharmaceutics (2012), 80 (2), 459-464CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)The aim of this study was to investigate whether amorphous indomethacin samples prepd. using different preparative techniques and processing parameters exhibit different structural and thermodn. characteristics and whether these differences can be correlated to their dissoln. behavior. Samples were prepd. either by cooling the drug melt at different cooling rates or by cryo-milling the drug for different milling times. The resulting amorphous materials were characterized using X-ray diffraction, Raman spectroscopy and polarising light microscopy. All samples were entirely X-ray amorphous, except for the sample cryo-milled for 15 min, which exhibited residual crystallinity. The shape of the halos in the diffractograms, however, varied depending on the prepn. method and processing parameters, suggesting structural variations in the near order of the mols. between the prepd. amorphous forms. This finding was supported by principal component anal. of the Raman spectra, as the samples clustered in the scores plot according to processing parameters for both of the preparative methods used. When investigating the dissoln. behavior, the samples cooled at different cooling rates showed no significant differences in their dissoln. profiles and dissoln. rates (≈0.55 μg/mL/cm2). In contrast, for cryo-milled samples, dissoln. rate depended on the milling time, with samples milled for 120, 180 and 240 min, showing significantly increased dissoln. rates of 0.28, 0.48 and 0.59 μg/mL/cm2, resp., when compared to cryst. indomethacin (≈0.06 and 0.05 μg/mL/cm2 for α and γ-indomethacin, resp.). The milling processes appear to continue to affect the degree of disorder in the solid material, enhancing its dissoln. rate, although all samples milled for >30 min were X-ray amorphous. Thus, choosing the right prepn. technique and parameters for prepg. amorphous solids is crit. for producing materials with enhanced dissoln. profiles.
- 27Alonzo, D. E.; Zhang, G. G. Z.; Zhou, D.; Gao, Y.; Taylor, L. S. Understanding the behavior of amorphous pharmaceutical systems during dissolution. Pharm. Res. 2010, 27 (4), 608– 618, DOI: 10.1007/s11095-009-0021-1Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhvV2gsLg%253D&md5=94caab8db7049285982b8a309dc579e9Understanding the Behavior of Amorphous Pharmaceutical Systems during DissolutionAlonzo, David E.; Zhang, Geoff G. Z.; Zhou, Deliang; Gao, Yi; Taylor, Lynne S.Pharmaceutical Research (2010), 27 (4), 608-618CODEN: PHREEB; ISSN:0724-8741. (Springer)Purpose: To investigate the underlying phys. processes taking place during dissoln. of amorphous pharmaceuticals and correlate them to the obsd. soln. concn.-time profiles. Felodipine and indomethacin were used as model hydrophobic compds. Concn.-time profiles were monitored during dissoln. of the model amorphous compds. using in situ fiber-optic UV spectroscopy. Crystn. of the solid exposed to an aq. environment was monitored using Raman spectroscopy and/or powder x-ray diffraction. Polarized light microscopy was used to provide qual. information about crystn. processes. For felodipine, a small extent of supersatn. was generated via dissoln. at 25°C but not at 37°C. The amorphous solid was found to crystallize rapidly at both temps. upon exposure to the dissoln. medium. Addn. of low concns. of polymers to the dissoln. medium was found to delay crystn. of the amorphous solid, leading to the generation of supersatd. solns. Amorphous indomethacin did not crystallize as readily in an aq. environment; hence, dissoln. resulted in supersatd. solns. However, crystn. from these supersatd. solns. was rapid. Polymeric additives were able to retard crystn. from supersatd. solns. of both indomethacin and felodipine for up to 4 h. The dissoln. advantage of amorphous solids can be negated either by crystn. of the amorphous solid on contact with the dissoln. medium or through rapid crystn. of the supersatd. soln. Polymeric additives can potentially retard both of these crystn. routes, leading to the generation of supersatd. solns. that can persist for biol. relevant timeframes.
- 28Mah, P. T.; Peltonen, L.; Novakovic, D.; Rades, T.; Strachan, C. J.; Laaksonen, T. The effect of surfactants on the dissolution behavior of amorphous formulations. Eur. J. Pharm. Biopharm. 2016, 103, 13– 22, DOI: 10.1016/j.ejpb.2016.03.007Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XltFGqtLk%253D&md5=e196ac6a38a5877f7d914e4e15ea4757The effect of surfactants on the dissolution behavior of amorphous formulationsMah, Pei T.; Peltonen, Leena; Novakovic, Dunja; Rades, Thomas; Strachan, Clare J.; Laaksonen, TimoEuropean Journal of Pharmaceutics and Biopharmaceutics (2016), 103 (), 13-22CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)The optimal design of oral amorphous formulations benefits from the use of excipients to maintain drug supersatn. and thus ensures adequate absorption during intestinal transit. The use of surfactants for the maintenance of supersatn. in amorphous formulations has not been investigated in detail. The main aim of this study was to investigate the effect of surfactant on the dissoln. behavior of neat amorphous drug and binary polymer based solid dispersion. Indomethacin was used as the model drug and the surfactants studied were polysorbate 80 and poloxamer 407. The presence of surfactants (alone or in combination with polymers) in the buffer was detrimental to the dissoln. of neat amorphous indomethacin, suggesting that the surfactants promoted the crystn. of neat amorphous indomethacin. In contrast, the presence of surfactants (0.01% w/v) in the buffer resulted in a significant improvement on the dissoln. behavior of binary polymer based solid dispersion. Incorporating the surfactant to the formulation to form ternary solid dispersion adversely affected the dissoln. behavior. In conclusion, the use of surfactants (as wetting or solubilization agents) in dissoln. studies of neat amorphous drugs requires prudent consideration. The design of amorphous formulations with optimal dissoln. performance requires the appropriate selection of a combination of excipients and consideration of the method of introducing the excipients.
- 29Karmwar, P.; Graeser, K.; Gordon, K. C.; Strachan, C. J.; Rades, T. Investigation of properties and recrystallisation behaviour of amorphous indomethacin samples prepared by different methods. Int. J. Pharm. 2011, 417 (1–2), 94– 100, DOI: 10.1016/j.ijpharm.2010.12.019Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVygsL3M&md5=c6177699d554012c2923f9d1d868e4d0Investigation of properties and recrystallisation behaviour of amorphous indomethacin samples prepared by different methodsKarmwar, Pranav; Graeser, Kirsten; Gordon, Keith C.; Strachan, Clare J.; Rades, ThomasInternational Journal of Pharmaceutics (2011), 417 (1-2), 94-100CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)The aim of this study was to investigate if amorphous indomethacin samples, prepd. using different prepn. methods, exhibit different structural and kinetic characteristics and if these differences can be correlated to their phys. stability (time to crystn.). Samples were prepd. by melt quenching, spray drying, ball milling, and cryo-milling. The resulting amorphous materials were characterized using X-ray diffraction, Raman spectroscopy and differential scanning calorimetry. All freshly prepd. samples were completely X-ray amorphous (with a halo being the only feature in the diffractograms). The shape of the halos in the diffractograms, however, varied depending on the prepn. method, suggesting structural variations in the near order of the mols. between the differently prepd. amorphous forms. Principal component anal. of the Raman spectra of the various amorphous forms revealed that the samples clustered in the scores plot according to prepn. method, again suggesting structural differences due to prepn. method. The range of vibrations assocd. with the largest spectral differences in the loadings plot showed that these differences were due to a range of mol. conformations and intermol. interactions. The ranking of the samples with respect to stability was: quench cooled amorphous samples > cryo-milled (α-form) > spray dried > ball milled (α-form) > ball milled (γ-form) = cryo-milled (γ-form). This ranking was not correlated with the diffractogram shapes or sample distribution in the scores plot of the Raman spectra, suggesting that phys. stability was not directly affected by structural variation in the samples. However, ranking of stability of the differently prepd. amorphous forms of the drug could be predicted by detg. the relaxation time values, for all amorphous samples. The relaxation times, calcd. by using the Adam Gibbs and Kohlrausch-Williams-Watts equations, were in accordance with the exptl. detd. stability order. This study showed that correlation of phys. stability with calcd. relaxation time is possible for the same amorphous systems prepd. by different methods. This could aid in selecting the most appropriate prepn. techniques in situations where there are a variety of suitable methods.
- 30Wu, T.; Yu, L. Surface crystallization of indomethacin below Tg. Pharm. Res. 2006, 23 (10), 2350– 2355, DOI: 10.1007/s11095-006-9023-4Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtVeqsbrF&md5=2c96d63050cd5154235bc4ecea6a3949Surface Crystallization of Indomethacin Below Glass Transition TemperatureWu, Tian; Yu, LianPharmaceutical Research (2006), 23 (10), 2350-2355CODEN: PHREEB; ISSN:0724-8741. (Springer)Purpose. To study the surface crystn. of indomethacin (IMC) below T g and its effects on the kinetics of overall crystn. Methods. Crystal growth rates in liq. layers formed between microscope cover glasses were measured with the top cover glass in place and removed. Polymorphs were identified by powder x-ray diffraction, Raman microscopy, and melting-point detn. by hot-stage microscopy. Surface crystals were identified by scratching the sample surface, by cutting the sample to expose its interior, and by analyzing the intensity of x-ray diffraction. Amorphous IMC particles of different sizes were stored at 40° (T g-2°) and analyzed at different times by differential scanning calorimetry to obtain the kinetics of crystn. Results. Crystal growth of IMC below T g at the free surface was approx. two orders of magnitude faster than that in the bulk, resulting in a surface layer of crystals around a slower-crystg. interior. Surface crystn. yielded mainly the γ polymorph. Amorphous IMC powders showed rapid initial crystn. at 40°, but the crystn. abruptly slowed down at "satn. levels" below 100%; the larger the particles, the lower the "satn. level." Conclusion. The faster surface crystn. of IMC than the bulk crystn. leads to unusual crystn. kinetics wherein a rapid initial increase of crystallinity is followed by an abrupt slowdown of crystn. Surface crystn. should be distinguished from bulk crystn. in modeling and controlling the crystn. of amorphous solids.
- 31Andronis, V.; Yoshioka, M.; Zografi, G. Effects of sorbed water on the crystallization of indomethacin from the amorphous state. J. Pharm. Sci. 1997, 86 (3), 346– 351, DOI: 10.1021/js9602711Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXhtVGhtrs%253D&md5=c5bdb49f1308a0812dc207822279dd12Effects of Sorbed Water on the Crystallization of Indomethacin from the Amorphous StateAndronis, Vlassios; Yoshioka, Minoru; Zografi, GeorgeJournal of Pharmaceutical Sciences (1997), 86 (3), 346-351CODEN: JPMSAE; ISSN:0022-3549. (American Chemical Society)The water sorption isotherm and the crystn. rates for amorphous indomethacin were detd. at 30° as a function of relative humidity (RH), along with the effects of water content on the glass transition temp. (Tg). Below 43% RH, only the stable γ crystal form appears, whereas at higher RH, only the metastable α crystal form appears. The tendency for the α crystals to form at higher RH is consistent with the Ostwald step rule. The crystn. rate of the α form continuously increased with increasing RH due to increasing mol. mobility. The crystn. mechanism of the γ form changed from surface-initiated to bulk-initiated crystn. at 21% RH, and although crystn. rates of the γ form increased with increasing RH in both cases, they were higher when crystn. was surface-initiated. The complex crystn. behavior of the γ form is explained by the higher water content and mol. mobility of the surface relative to the bulk and the general effect of water on α or γ crystal form selection as described by the Ostwald step rule.
- 32Priemel, P. A.; Grohganz, H.; Gordon, K. C.; Rades, T.; Strachan, C. J. The impact of surface- and nano-crystallisation on the detected amorphous content and the dissolution behaviour of amorphous indomethacin. Eur. J. Pharm. Biopharm. 2012, 82 (1), 187– 193, DOI: 10.1016/j.ejpb.2012.05.012Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtV2mtLnL&md5=9c9d7f4b2578d1d064d66cf4691b0945The impact of surface- and nano-crystallisation on the detected amorphous content and the dissolution behaviour of amorphous indomethacinPriemel, P. A.; Grohganz, H.; Gordon, K. C.; Rades, T.; Strachan, C. J.European Journal of Pharmaceutics and Biopharmaceutics (2012), 82 (1), 187-193CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)The crystallinity and phys. stability of amorphous drugs has previously been studied using different anal. techniques. However, the effect of the measurement method on obsd. crystallinity and its importance for crit. quality attributes, such as dissoln., has not yet been widely investigated.The aim of this study was to (i) qual. analyze and understand the recrystn. behavior of amorphous indomethacin during storage, (ii) quantify the amorphous content during storage with complementary anal. techniques and (iii) investigate the relationship between obsd. recrystn. behavior and dissoln. behavior.Quench cooled indomethacin was stored and the samples were visualized by SEM to gain spatially resolved information about the recrystn. behavior. Crystn. was quantified by Fourier transform attenuated total reflectance IR (FT-ATR-IR) spectroscopy, differential scanning calorimetry and X-ray powder diffraction.These techniques resulted in different obsd. recrystn. profiles. The physicochem. phenomena detected and sampling geometry for each technique together with the sample recrystg. from the surface and appearance of nano-crystals were used to explain the differences.The dissoln. behavior at the obsd. recrystn. endpoints for the different anal. techniques revealed that FT-ATR-IR spectroscopy predicted the changes in dissoln. behavior due to crystn. best.
- 33Tres, F.; Treacher, K.; Booth, J.; Hughes, L. P.; Wren, S. A. C.; Aylott, J. W.; Burley, J. C. Indomethacin-Kollidon VA64 extrudates: A mechanistic study of pH-dependent controlled release. Mol. Pharmaceutics 2016, 13 (3), 1166– 1175, DOI: 10.1021/acs.molpharmaceut.5b00979Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvV2rs7k%253D&md5=9cbf9d0af9b46777e849b7ccc6e8b36cIndomethacin-Kollidon VA64 Extrudates: A Mechanistic Study of pH-Dependent Controlled ReleaseTres, Francesco; Treacher, Kevin; Booth, Jonathan; Hughes, Leslie P.; Wren, Stephen A. C.; Aylott, Jonathan W.; Burley, Jonathan C.Molecular Pharmaceutics (2016), 13 (3), 1166-1175CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Because of its weakly acidic nature (pKa of 4.5), indomethacin presents an aq. soly. that significantly increases when changing from acidic to neutral/alk. pH (1.5 μg/mL at pH 1.2 and 105.2 μg/mL at pH 7.4). We have therefore investigated the impact of the dissoln. medium pH on the dissoln. performance of indomethacin:Kollidon VA64 extrudates. The impact of the drug loading on the dissoln. properties of these systems was also examd. (5%, 15%, 30%, 50%, 70%, and 90% drug loading). Time-resolved Raman spectroscopy along with in-line UV-vis spectrophotometry was employed to directly relate changes in dissoln. behavior to physicochem. changes that occur to the extrudate during the test. The dissoln. tests were performed in pH 2 HCl (to mimic the stomach conditions), and this was then switched during the expt. to pH 6.8 phosphate buffer (to simulate the poststomach conditions). The rotating disk dissoln. rate test was also used to simultaneously measure the dissoln. rate of both the drug and the polymer. We found that in pH 2 HCl buffer, for the 15% or higher drug-loaded extrudates, Kollidon VA64 preferentially dissolves from the exterior of the compact leaving an amorphous drug-rich hydrophobic shell, which, similarly to an enteric coating, inhibits the drug release. The in situ formation of an enteric coating has been previously hypothesized, and this has been the first time that is directly obsd. in a pH-variable dissoln. test. The dissoln. medium switch to pH 6.8 phosphate buffer, due to the large increase of the aq. soly. of indomethacin at this pH, leads to rapid dissoln. of the material forming the coating and therefore total drug release. In contrast, the 5% extrudate is fully hydrated and quickly dissolves at low pH pointing to a dissoln. performance dependent on highly water-sol. Kollidon VA64.
- 34Koranne, S.; Thakral, S.; Suryanarayanan, R. Effect of formulation and process parameters on the disproportionation of indomethacin sodium in buffered lyophilized formulations. Pharm. Res. 2018, 35 (11), 214, DOI: 10.1007/s11095-018-2499-xGoogle ScholarThere is no corresponding record for this reference.
- 35Peltonen, L.; Liljeroth, P.; Heikkilä, T.; Kontturi, K.; Hirvonen, J. Dissolution testing of acetylsalicylic acid by a channel flow method—correlation to USP basket and intrinsic dissolution methods. Eur. J. Pharm. Sci. 2003, 19 (5), 395– 401, DOI: 10.1016/S0928-0987(03)00140-4Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXlvFyiurY%253D&md5=6df5aa626ee4587f80bd35328bb4bf29Dissolution testing of acetylsalicylic acid by a channel flow method-correlation to USP basket and intrinsic dissolution methodsPeltonen, Leena; Liljeroth, Peter; Heikkila, Tiina; Kontturi, Kyosti; Hirvonen, JouniEuropean Journal of Pharmaceutical Sciences (2003), 19 (5), 395-401CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)A new modification of the channel flow dissoln. method is introduced together with the theor. basis to ext. the soly. and mass transfer parameters from the dissoln. expts. Correlation of drug dissoln. profiles in the channel flow app. was evaluated with respect to USP basket and intrinsic dissoln. methods at pH 1.2 or 6.8. Acetylsalicylic acid (ASA) was studied as a pure drug substance and as three simple tablet compns. with microcryst. cellulose (MCC) and/or lactose as excipients. The channel flow measurements of 100% ASA tablets correlated well with the results of intrinsic dissoln. tests. In the channel flow method as well as in the USP basket method the release of ASA was fastest from the tablet compns. contg. lactose, while the slowest dissoln. rate was obsd. with the compn. contg. MCC as the only excipient. As presumed, the dissoln. rate of the weak acid was decreased as the pH of the medium was lowered, which was clearly confirmed also by the three dissoln. methods. MCC forms matrix tablets and in the USP basket method the dissoln. profiles followed square root of time kinetics indicating that diffusion was the rate-controlling step of ASA dissoln. Also the channel flow results indicated that the dissoln. Of ASA was controlled by mass transfer. The swelling behavior of the tablets is different in the channel flow method as compared to the basket method: only one tablet surface is exposed to the dissoln. medium in the channel flow system. The contact between the tablet surface and the dissoln. medium is more similar between the channel flow and intrinsic dissoln. methods.
- 36Sarnes, A.; Østergaard, J.; Jensen, S. S.; Aaltonen, J.; Rantanen, J.; Hirvonen, J.; Peltonen, L. Dissolution study of nanocrystal powders of a poorly soluble drug by UV imaging and channel flow methods. Eur. J. Pharm. Sci. 2013, 50 (3), 511– 519, DOI: 10.1016/j.ejps.2013.08.030Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFCitLvK&md5=ef415dcf6bbdd663d96b5e9d9693813aDissolution study of nanocrystal powders of a poorly soluble drug by UV imaging and channel flow methodsSarnes, Annika; Oestergaard, Jesper; Jensen, Sabrine Smedegaard; Aaltonen, Jaakko; Rantanen, Jukka; Hirvonen, Jouni; Peltonen, LeenaEuropean Journal of Pharmaceutical Sciences (2013), 50 (3-4), 511-519CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)Application of drug nanocrystals provides advantageous options for the pharmaceutical formulation development of poorly sol. drugs. The objective of this study was to investigate the dissoln. behavior improving effects of differently sized nanocrystals of a poorly sol. model drug, indomethacin. Nanocrystal suspensions were prepd. using a top-down wet milling technique with three stabilizers: poloxamer F68, poloxamer F127 and polysorbate 80. The dissoln. of the differently sized indomethacin nanocrystals were investigated using a channel flow dissoln. method and by UV imaging. Unmilled bulk indomethacin and phys. mixts. were used as refs. According to both the dissoln. methods, the dissoln. properties of indomethacin were improved by the particle size redn. UV imaging was used for the first time as a dissoln. testing method for fast dissolving nanoscale material. The technique provided new information about the concn. of the dissolved drug next to the sample surface; with the smallest nanocrystals (580 nm) the indomethacin concn. next to the particle surface exceeded five-fold the thermodn. satd. indomethacin soln. concn. Thus the soly. improvement itself, not only the increased surface area for dissoln., may have an important role in the higher dissoln. rates of nanocrystal formulations. Poloxamer F68 was the most optimal stabilizer in the prepn. of the indomethacin nanocrystal suspensions and in the soly. and dissoln. enhancement as well.
- 37Sunesen, V. H.; Pedersen, B. L.; Kristensen, H. G.; Müllertz, A. In vivo in vitro correlations for a poorly soluble drug, danazol, using the flow-through dissolution method with biorelevant dissolution media. Eur. J. Pharm. Sci. 2005, 24 (4), 305– 313, DOI: 10.1016/j.ejps.2004.11.007Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhslWmsLc%253D&md5=e3fca526cc00dffbeadd9b1c856eb658In vivo in vitro correlations for a poorly soluble drug, danazol, using the flow-through dissolution method with biorelevant dissolution mediaSunesen, Vibeke Hougaard; Pedersen, Betty Lomstein; Kristensen, Henning Gjelstrup; Muellertz, AnetteEuropean Journal of Pharmaceutical Sciences (2005), 24 (4), 305-313CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)The purpose of the study was to design dissoln. tests that were able to distinguish between the behavior of danazol under fasted and fed conditions, by biorelevant media. In vitro dissoln. of 100 mg danazol capsules was performed using the flow-through dissoln. method. Flow rates were 8, 16 or 32 mL/min, corresponding to total vols. dissoln. medium of 960, 1920 and 3840 mL, resp. The media used contained bile salt and phospholipid levels relevant for either fasted or fed conditions in vivo. Crude and inexpensive bile components, Porcine Bile Ext. and soybean phospholipids, were used as the bile source. The effect of adding different concns. and molar ratios of monoglycerides and fatty acids to the fed state media was investigated. In vivo release profiles under fasted and fed conditions were obtained from a previous study by deconvolution [Sunesen, V.H., Vedelsdal, R., Kristensen, H.G., Christrup, L., Muellertz, A. 2005]. Effect of liq. vol. and food intake on the abs. bioavailability of danazol, a poorly sol. drug, [Eur. J. Pharm. Sci. 24, 297-303]. In the fasted state, the physiol. most relevant correlation with in vivo results was achieved with a medium contg. 6.3 mM bile salts and 1.25 mM phospholipids (8 mL/min). A medium contg. 18.8 mM bile salts, 3.75 mM phospholipids, 4.0 mM monoglycerides and 30 mM fatty acids (8 mL/min) gave the closest correlation with fed state in vivo results. By using the flow-through dissoln. method it was possible to obtain correlations with in vivo release of danazol under fasted and fed conditions. Both hydrodynamics and medium compn. were important for the dissoln. of danazol. In the fed state an IVIVC could only be obtained by including monoglycerides and fatty acids in the medium.
- 38Greco, K.; Bogner, R. Solution-mediated phase transformation: Significance during dissolution and implications for bioavailability. J. Pharm. Sci. 2012, 101 (9), 2996– 3018, DOI: 10.1002/jps.23025Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1OltbnF&md5=f94251f8c92af8013a346b316823bf4fSolution-mediated phase transformation: Significance during dissolution and implications for bioavailabilityGreco, Kristyn; Bogner, RobinJournal of Pharmaceutical Sciences (2012), 101 (9), 2996-3018CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)A review. Soly. improvement of poorly sol. drug compds. is a key approach to ensuring the successful development of many new drugs. Methods used to improve the soly. of drug compds. include forming a salt, cocrystal, or amorphous solid. These methods of improving soly. can often lead to a phenomenon called soln.-mediated phase transformation, a phase change that is facilitated through exposure to soln. Soln.-mediated phase transformation occurs in three steps: dissoln. to create a supersatd. soln. followed by nucleation of less sol. phase and the growth of that phase. When the growth of the less sol. phase occurs on the surface of the metastable solid, this phenomenon can cause a marked decrease in dissoln. rate during in vitro dissoln. evaluation, and ultimately in vivo. Therefore, transformation to a less sol. solid during dissoln. is an important aspect to consider when evaluating approaches to increase the soly. of a poorly sol. drug. Identification of soln.-mediated phase transformation during dissoln. is reviewed for powder dissoln., rotating disk method, and channel flow-through app. Types of soln.-mediated phase transformation are described in this report, including those involving salts, polymorphs, amorphous solids, and cocrystals. Many exptl. examples are provided. Evidence of potential soln.-mediated phase transformation in vivo is discussed to better understand the relationship between in vitro dissoln. evaluation and in vivo performance. © 2011 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 39Slavin, P. A.; Sheen, D. B.; Shepherd, E. E. A.; Sherwood, J. N.; Feeder, N.; Docherty, R.; Milojevic, S. Morphological evaluation of the γ-polymorph of indomethacin. J. Cryst. Growth 2002, 237–239, 300– 305, DOI: 10.1016/S0022-0248(01)01924-8Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XjvVSktr8%253D&md5=ecfc82d311655b5cc96384253b4eef3aMorphological evaluation of the γ-polymorph of indomethacinSlavin, Paul A.; Sheen, David B.; Shepherd, Evelyn E. A.; Sherwood, John N.; Feeder, Neil; Docherty, Robert; Milojevic, SnezenaJournal of Crystal Growth (2002), 237-239 (Pt. 1), 300-305CODEN: JCRGAE; ISSN:0022-0248. (Elsevier Science B.V.)A study was made of the polymorphic nature of crystals of the pharmaceutical indomethacin grown from a wide range of solvents and from the melt. In most solvents, growth at high supersaturations yielded either a 1:0.5 solvated form (approx.) or the α-polymorph. At low supersaturations the γ-polymorph was commonly produced. Solns. in MeOH and tBuOH yielded a 1:1 solvate. The morphol. of the γ-form showed no variation with solvent type but changed with supersatn. in a manner consistent with a differential variation in growth rates of the faces. This lack of solvent influence was confirmed by the fact that a similar morphol. resulted on growth from the melt. Morphol. predictions were carried out for the γ-polymorph and these show good agreement with exptl. observations.
- 40Hartshorn, C. M.; Lee, Y. J.; Camp, C. H.; Liu, Z.; Heddleston, J.; Canfield, N.; Rhodes, T. A.; Hight Walker, A. R.; Marsac, P. J.; Cicerone, M. T. Multicomponent chemical imaging of pharmaceutical solid dosage forms with broadband CARS microscopy. Anal. Chem. 2013, 85 (17), 8102– 8111, DOI: 10.1021/ac400671pGoogle ScholarThere is no corresponding record for this reference.
- 41Taylor, L. S.; Zografi, G. Spectroscopic characterization of interactions between PVP and indomethacin in amorphous molecular dispersions. Pharm. Res. 1997, 14 (12), 1691– 1698, DOI: 10.1023/A:1012167410376Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXktFKjtQ%253D%253D&md5=9bc968c8777589f6d0d90b2f349237ddSpectroscopic characterization of interactions between PVP and indomethacin in amorphous molecular dispersionsTaylor, Lynne S.; Zografi, GeorgePharmaceutical Research (1997), 14 (12), 1691-1698CODEN: PHREEB; ISSN:0724-8741. (Plenum Publishing Corp.)The mol. structure of indomethacin-PVP amorphous solid dispersions was studied and specific interactions between the components was identified using vibrational spectroscopy. Solid dispersions of PVP and indomethacin were prepd. using a solvent evapn. technique and IR and FT-Raman spectra were obtained. A comparison of the carbonyl stretching region of γ indomethacin, known to form carboxylic acid dimers, with that of amorphous indomethacin indicated that the amorphous phase exists predominantly as dimers. The hydrogen bonding of α indomethacin is not as dimers. Addn. of PVP to amorphous indomethacin increased the intensity of the IR band assigned to non-hydrogen bonded carbonyl. Concomitantly, the PVP carbonyl stretch appeared at a lower wavenumber indicating hydrogen bonding. Model solvent systems aided spectral interpretation. The magnitude of the spectral changes were comparable for an indomethacin-PVP solid dispersion and a soln. of indomethacin in methylpyrrolidone at the same wt. percent. Indomethacin interacts with PVP in solid dispersions through hydrogen bonds formed between the drug hydroxyl and polymer carbonyl resulting in disruption of indomethacin dimers. PVP may influence the crystn. kinetics by preventing the self assocn. of indomethacin mols. The similarity of results for solid dispersions and solns. emphasizes the "soln." nature of this binary amorphous state.
- 42Trasi, N. S.; Purohit, H. S.; Taylor, L. S. Evaluation of the crystallization tendency of commercially available amorphous tacrolimus formulations exposed to different stress conditions. Pharm. Res. 2017, 34 (10), 2142– 2155, DOI: 10.1007/s11095-017-2221-4Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFCrsbvP&md5=4d1019be54fe12ef75f30251a14f6869Evaluation of the Crystallization Tendency of Commercially Available Amorphous Tacrolimus Formulations Exposed to Different Stress ConditionsTrasi, Niraj S.; Purohit, Hitesh S.; Taylor, Lynne S.Pharmaceutical Research (2017), 34 (10), 2142-2155CODEN: PHREEB; ISSN:0724-8741. (Springer)Tacrolimus, an immunosuppressant, is a poorly water sol. compd. whereby the com. available capsule formulations contain the drug in amorphous form. The goal of this study was to evaluate the robustness of the innovator product and five generic formulations to crystn. following storage at stress conditions. Methods: Products were purchased from a pharmacy and stored at 40/75% relative humidity (RH), open dish conditions. Crystallinity was detd. using x-ray diffraction. The quantity of the ingredients in the formulations were detd. using different approaches and the various factors that might cause instability in the formulations were studied. Results: After 4 wk of open dish storage at 40/75% RH, one of the generic formulations showed evidence of tacrolimus crystn. Further investigations revealed batch-to-batch variations in crystn. tendency with the extent of crystallinity varying between 50 and 100% for different batches. Crystn. was also obsd. at lower storage temps. (30) when the RH was maintained at 75%. It was found that crystn. could be induced in a model formulation by wet granulating an ethanolic soln. of the drug with lactose and drying at 60-70 followed by exposure to stress conditions. Conclusions: It seems probable that the generic that was susceptible to crystn. contains amorphous drug phys. mixed with polymeric excipients, rather than as an amorphous solid dispersion. This study highlights the importance of considering the manufg. process on the stability of the resultant amorphous product.
- 43Sun, D. D.; Lee, P. I. Evolution of supersaturation of amorphous pharmaceuticals: The effect of rate of supersaturation generation. Mol. Pharmaceutics 2013, 10 (11), 4330– 4346, DOI: 10.1021/mp400439qGoogle Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFSms7%252FO&md5=d7452e8a147c4f673223032871751f51Evolution of Supersaturation of Amorphous Pharmaceuticals: The Effect of Rate of Supersaturation GenerationSun, Dajun D.; Lee, Ping I.Molecular Pharmaceutics (2013), 10 (11), 4330-4346CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The combination of a rapidly dissolving and supersaturating spring with a pptn. retarding parachute has often been pursued as an effective formulation strategy for amorphous solid dispersions (ASDs) to enhance the rate and extent of oral absorption. However, the interplay between these two rate processes in achieving and maintaining supersatn. remains inadequately understood, and the effect of rate of supersatn. buildup on the overall time evolution of supersatn. during the dissoln. of amorphous solids has not been explored. The objective of this study is to investigate the effect of supersatn. generation rate on the resulting kinetic soly. profiles of amorphous pharmaceuticals and to delineate the evolution of supersatn. from a mechanistic viewpoint. Exptl. concn.-time curves under varying rates of supersatn. generation and recrystn. for model drugs, indomethacin (IND), naproxen (NAP) and piroxicam (PIR), were generated from infusing dissolved drug (e.g., in ethanol) into the dissoln. medium and compared with that predicted from a comprehensive mechanistic model based on the classical nucleation theory taking into account both the particle growth and ripening processes. In the absence of any dissolved polymer to inhibit drug pptn., both our exptl. and predicted results show that the max. achievable supersatn. (i.e., kinetic soly.) of the amorphous solids increases, the time to reach max. decreases, and the rate of concn. decline in the de-supersatn. phase increases, with increasing rate of supersatn. generation (i.e., dissoln. rate). Our mechanistic model also predicts the existence of an optimal supersatn. rate which maximizes the area under the curve (AUC) of the kinetic soly. concn.-time profile, which agrees well with exptl. data. In the presence of a dissolved polymer from ASD dissoln., these obsd. trends also hold true except the de-supersatn. phase is more extended due to the crystn. inhibition effect. Since the obsd. kinetic soly. of nonequil. amorphous solids depends on the rate of supersatn. generation, our results also highlight the underlying difficulty in detg. a reproducible soly. advantage for amorphous solids.
- 44Zhu, L.; Wong, L.; Yu, L. Surface-enhanced crystallization of amorphous nifedipine. Mol. Pharmaceutics 2008, 5 (6), 921– 926, DOI: 10.1021/mp8000638Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtFChsbvE&md5=9f87dc51ab6f12c143b2760fa719346bSurface-Enhanced Crystallization of Amorphous NifedipineZhu, Lei; Wong, Letitia; Yu, LianMolecular Pharmaceutics (2008), 5 (6), 921-926CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Amorphous solids are generally more sol. and faster dissolving than their cryst. counterparts, a property useful for delivering poorly sol. drugs. Amorphous drugs must be stable against crystn., for crystn. negates their advantages. Recent studies found that crystal growth in amorphous indomethacin is orders of magnitude faster at the free surface than through the bulk and this surface-enhanced crystn. can be inhibited by an ultrathin coating. Herein, we report a second system that exhibits the same phenomena. Crystal growth at the free surface of amorphous nifedipine (NIF) was at least 1 order of magnitude faster than that through the bulk below the glass transition temp. Tg (42 °C). A thin coating of gold (10 nm) reduced the surface crystal growth rate to the bulk crystal growth rate. Surface-enhanced crystal growth was more pronounced near and below Tg than substantially above Tg, which suggests that this growth mode is more important for the glassy state. Our results support the view that a thin layer of mols. near the surface have higher mobility than the bulk mols. and can enable faster crystal growth. The higher mobility of surface mols. and the resulting fast crystal growth can be suppressed by an ultrathin coating.
- 45Hasebe, M.; Musumeci, D.; Yu, L. Fast surface crystallization of molecular glasses: creation of depletion zones by surface diffusion and crystallization flux. J. Phys. Chem. B 2015, 119 (7), 3304– 3311, DOI: 10.1021/jp512400cGoogle Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlyksro%253D&md5=893b871632f9b82d57a79d5140c69c47Fast Surface Crystallization of Molecular Glasses: Creation of Depletion Zones by Surface Diffusion and Crystallization FluxHasebe, Mariko; Musumeci, Daniele; Yu, LianJournal of Physical Chemistry B (2015), 119 (7), 3304-3311CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Mol. glasses can grow crystals much faster at the free surface than in the interior. A property of this process is the creation of depressed grooves or depletion zones around the crystals on the initially flat amorphous surface. With SEM and at. force microscopy, the authors studied this phenomenon in indomethacin (IMC), which crystallizes in two polymorphs (α and γ) of different morphologies. The obsd. depletion zones are well reproduced by the known coeffs. of surface diffusion and the velocities of crystal growth. At the slow-growing flanks of needle-like α-IMC crystals, depletion zones widen and deepen over time according to the expected kinetics for surface diffusion responding to a crystn. flux. Before fast-advancing growth fronts, depletion zones have less time to develop; their steady-state dimensions agree with the same model revised for a moving phase boundary. These results support the view that surface diffusion enables fast surface crystal growth on mol. glasses. The authors' finding helps understand crystal growth in thin films in which the formation of deep depletion zones can cause dewetting and alter growth kinetics.
- 46Zhu, L.; Jona, J.; Nagapudi, K.; Wu, T. Fast surface crystallization of amorphous griseofulvin below Tg. Pharm. Res. 2010, 27 (8), 1558– 1567, DOI: 10.1007/s11095-010-0140-8Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXltV2ksr8%253D&md5=e3740687002f83f51e2acfa5b5c7e759Fast Surface Crystallization of Amorphous Griseofulvin Below TgZhu, Lei; Jona, Janan; Nagapudi, Karthik; Wu, TianPharmaceutical Research (2010), 27 (8), 1558-1567CODEN: PHREEB; ISSN:0724-8741. (Springer)To study crystal growth rates of amorphous griseofulvin (GSF) below its glass transition temp. (Tg) and the effect of surface crystn. on the overall crystn. kinetics of amorphous GSF. Amorphous GSF was generated by melt quenching. Surface and bulk crystal growth rates were detd. using polarized light microscope. X-ray powder diffraction (XRPD) and Raman microscopy were used to identify the polymorph of the crystals. Crystn. kinetics of amorphous GSF powder stored at 40° (Tg-48°) and room temp. (Tg-66°) was monitored using XRPD. Results: Crystal growth at the surface of amorphous GSF is 10- to 100-fold faster than that in the bulk. The surface crystal growth can be suppressed by an ultrathin gold coating. Below Tg, the crystn. of amorphous GSF powder was biphasic with a rapid initial crystn. stage dominated by the surface crystn. and a slow or suspended late stage controlled by the bulk crystn. GSF exhibits the fastest surface crystn. kinetics among the known amorphous pharmaceutical solids. Well below Tg, surface crystn. dominated the overall crystn. kinetics of amorphous GSF powder. Thus, surface crystn. should be distinguished from bulk crystn. in studying, modeling and controlling the crystn. of amorphous solids.
- 47Elkhabaz, A.; Sarkar, S.; Dinh, J. K.; Simpson, G. J.; Taylor, L. S. Variation in supersaturation and phase behavior of ezetimibe amorphous solid dispersions upon dissolution in different biorelevant media. Mol. Pharmaceutics 2018, 15 (1), 193– 206, DOI: 10.1021/acs.molpharmaceut.7b00814Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvVeisrvK&md5=fac144655adaafc768cbe00785514451Variation in Supersaturation and Phase Behavior of Ezetimibe Amorphous Solid Dispersions upon Dissolution in Different Biorelevant MediaElkhabaz, Ahmed; Sarkar, Sreya; Dinh, Janny K.; Simpson, Garth J.; Taylor, Lynne S.Molecular Pharmaceutics (2018), 15 (1), 193-206CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The delivery of poorly water-sol. drugs using amorphous solid dispersions (ASDs) has been widely acknowledged as a promising strategy for enhancing oral bioavailability. Upon dissoln., ASDs have accelerated dissoln. rates and yield supersatd. solns. leading to higher apparent solubilities. Understanding the complex phase behavior of ASDs during dissoln. is crucial for developing an effective formulation. Since the absorption of a lipophilic, high permeability drug is detd. primarily by the intraluminal dissoln. process and the final concn. achieved, there is a need for evaluation in biorelevant dissoln. media that simulate both fasting and fed gastrointestinal states. In this study, using ezetimibe as a model drug, three different ASDs were prepd. using poly(acrylic acid) (PAA), polyvinylpyrrolidone (PVP), and hydroxypropyl methylcellulose acetyl succinate (HPMC-AS). Dissoln. of ASDs was carried out in sodium phosphate buffer, fed-state simulated intestinal fluid (FeSSIF), and Ensure Plus to evaluate the impact of different dissoln. media on release profile, supersatn., and phase behavior. The supersatn. level and crystn. kinetics varied among the dispersions and were found to be highly dependent on the medium employed. The presence of solubilizing additives in biorelevant media greatly affected the generation and stabilization of supersatd. solns. Second harmonic generation microscopy was found to enable the detection of crystals in all media including the highly turbid Ensure Plus system. In conclusion, it is important to evaluate the impact of complex biorelevant media on the dissoln. performance of ASDs to better design supersaturating formulations for oral delivery.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 31 publications.
- Alba M. Arbiol Enguita, Laurin Zöller, Teemu Tomberg, Mikko J. Heikkilä, Jukka K. S. Saarinen, Lea Wurr, Tom Konings, Alexandra Correia, Christoph Saal, Jennifer Dressman, Clare J. Strachan. Chemically Specific Coherent Raman Imaging of Liquid–Liquid Phase Separation and Its Sequelae. Analytical Chemistry 2025, 97
(6)
, 3242-3252. https://doi.org/10.1021/acs.analchem.4c03923
- Teemu Tomberg, Ilona Hämäläinen, Clare J. Strachan, Bert van Veen. Dynamic Phase Behavior of Amorphous Solid Dispersions Revealed with In Situ Stimulated Raman Scattering Microscopy. Molecular Pharmaceutics 2024, 21
(12)
, 6444-6457. https://doi.org/10.1021/acs.molpharmaceut.4c01032
- Filip Hládek, David Zůza, Ondřej Navrátil, Jan Tomas, Aleš Zadražil, Vladimír Novák, František Štěpánek. Evaluation of Individual and Crystal Population Dissolution Rates by Time-Resolved X-ray Microtomography. Crystal Growth & Design 2024, 24
(13)
, 5468-5477. https://doi.org/10.1021/acs.cgd.4c00113
- Malik Salman Haider, Arun Kumar Mahato, Anastasiia Kotliarova, Stefan Forster, Bettina Böttcher, Philipp Stahlhut, Yulia Sidorova, Robert Luxenhofer. Biological Activity In Vitro, Absorption, BBB Penetration, and Tolerability of Nanoformulation of BT44:RET Agonist with Disease-Modifying Potential for the Treatment of Neurodegeneration. Biomacromolecules 2023, 24
(10)
, 4348-4365. https://doi.org/10.1021/acs.biomac.2c00761
- Jaana Koskela, Joshua J. Sutton, Tiina Lipiäinen, Keith C. Gordon, Clare J. Strachan, Sara J. Fraser-Miller. Low- versus Mid-frequency Raman Spectroscopy for in Situ Analysis of Crystallization in Slurries. Molecular Pharmaceutics 2022, 19
(7)
, 2316-2326. https://doi.org/10.1021/acs.molpharmaceut.2c00126
- Ka̅rlis Be̅rziņš, Sara J. Fraser-Miller, Rong Di, Jingwen Liu, Leena Peltonen, Clare J. Strachan, Thomas Rades, Keith C. Gordon. Combined Effect of the Preparation Method and Compression on the Physical Stability and Dissolution Behavior of Melt-Quenched Amorphous Celecoxib. Molecular Pharmaceutics 2021, 18
(3)
, 1408-1418. https://doi.org/10.1021/acs.molpharmaceut.0c01208
- Dunja Novakovic, Leena Peltonen, Antti Isomäki, Sara J. Fraser-Miller, Line Hagner Nielsen, Timo Laaksonen, Clare J. Strachan. Surface Stabilization and Dissolution Rate Improvement of Amorphous Compacts with Thin Polymer Coatings: Can We Have It All?. Molecular Pharmaceutics 2020, 17
(4)
, 1248-1260. https://doi.org/10.1021/acs.molpharmaceut.9b01263
- Eman M. Mohamed, Mariame A. Hassan, Gereziher Sibhat, Tahir Khuroo, Ziyaur Rahman, Mansoor A. Khan. Effect of patients in-use and accelerated stability conditions on quality attributes and pharmacokinetic profile of four FDA approved extended-release anti-epileptic-drug products. International Journal of Pharmaceutics 2025, 668 , 124840. https://doi.org/10.1016/j.ijpharm.2024.124840
- Luchi Li, Qonita Kurnia Anjani, Aaron R. J. Hutton, Mingshan Li, Akmal Hidayat Bin Sabri, Lalitkumar Vora, Yara A. Naser, Yushi Tao, Helen O. McCarthy, Ryan F. Donnelly. Evaluation of physical and chemical modifications to drug reservoirs for stimuli-responsive microneedles. Drug Delivery and Translational Research 2024, 190 https://doi.org/10.1007/s13346-024-01737-0
- Roman Svoboda, Nicola Koutná, Magdalena Hynková, Marek Pakosta. In Situ Raman Spectroscopy as a Valuable Tool for Monitoring Crystallization Kinetics in Molecular Glasses. Molecules 2024, 29
(19)
, 4769. https://doi.org/10.3390/molecules29194769
- Stefanie Walter, Paulo G. M. Mileo, Mohammad Atif Faiz Afzal, Samuel O. Kyeremateng, Matthias Degenhardt, Andrea R. Browning, John C. Shelley. Predicting the Release Mechanism of Amorphous Solid Dispersions: A Combination of Thermodynamic Modeling and In Silico Molecular Simulation. Pharmaceutics 2024, 16
(10)
, 1292. https://doi.org/10.3390/pharmaceutics16101292
- Aleksandra Rzewińska, Jakub Szlęk, Damian Dąbrowski, Ewelina Juszczyk, Katarzyna Mróz, Heikki Räikkönen, Mia Siven, Maciej Wieczorek, Przemysław Dorożyński. Development of a Formulation and In Vitro Evaluation of a Pulmonary Drug Delivery System for a Novel Janus Kinase (JAK) Inhibitor, CPL409116. Pharmaceutics 2024, 16
(9)
, 1157. https://doi.org/10.3390/pharmaceutics16091157
- Israa Saad Mustafa Ali, Uday Aziz Sajad, Bazigha K. Abdul Rasool, . Solid dispersion systems for enhanced dissolution of poorly water-soluble candesartan cilexetil: In vitro evaluation and simulated pharmacokinetics studies. PLOS ONE 2024, 19
(6)
, e0303900. https://doi.org/10.1371/journal.pone.0303900
- Jukka Rantanen, Thomas Rades, Clare Strachan. Solid-state analysis for pharmaceuticals: Pathways to feasible and meaningful analysis. Journal of Pharmaceutical and Biomedical Analysis 2023, 236 , 115649. https://doi.org/10.1016/j.jpba.2023.115649
- Teemu Tomberg, Alba Maria Arbiol Enguita, Clare Strachan. Insights into solid dosage forms with nonlinear optical imaging. Journal of Medical Science 2023, 92
(3)
, e914. https://doi.org/10.20883/medical.e914
- Stefanie Dohrn, Samuel O. Kyeremateng, Esther Bochmann, Ekaterina Sobich, Andrea Wahl, Bernd Liepold, Gabriele Sadowski, Matthias Degenhardt. Thermodynamic Modeling of the Amorphous Solid Dispersion-Water Interfacial Layer and Its Impact on the Release Mechanism. Pharmaceutics 2023, 15
(5)
, 1539. https://doi.org/10.3390/pharmaceutics15051539
- Manjot Kaur, Deenan Santhiya. Fabrication of soy film with in-situ mineralized bioactive glass as a functional food for bone health. Food Bioscience 2022, 47 , 101767. https://doi.org/10.1016/j.fbio.2022.101767
- Olivier Jennotte, Nathan Koch, Anna Lechanteur, Brigitte Evrard. Development of amorphous solid dispersions of cannabidiol: Influence of the carrier, the hot-melt extrusion parameters and the use of a crystallization inhibitor. Journal of Drug Delivery Science and Technology 2022, 71 , 103372. https://doi.org/10.1016/j.jddst.2022.103372
- Jie Ren, Shijie Mao, Jidong Lin, Ying Xu, Qiaoqiao Zhu, Ning Xu. Research Progress of Raman Spectroscopy and Raman Imaging in Pharmaceutical
Analysis. Current Pharmaceutical Design 2022, 28
(18)
, 1445-1456. https://doi.org/10.2174/1381612828666220518145635
- Alexander P. Voronin, Nikita A. Vasilev, Artem O. Surov, Andrei V. Churakov, German L. Perlovich. Exploring the solid form landscape of the antifungal drug isavuconazole: crystal structure analysis, phase transformation behavior and dissolution performance. CrystEngComm 2021, 23
(48)
, 8513-8526. https://doi.org/10.1039/D1CE01353J
- Ahmet R. Dok, Thibaut Legat, Yovan de Coene, M. A. van der Veen, T. Verbiest, Stijn Van Cleuvenbergen. Nonlinear optical probes of nucleation and crystal growth: recent progress and future prospects. Journal of Materials Chemistry C 2021, 9
(35)
, 11553-11568. https://doi.org/10.1039/D1TC02007B
- Alex M. Sherman, Nita Takanti, Jiayue Rong, Garth J. Simpson. Nonlinear optical characterization of pharmaceutical formulations. TrAC Trends in Analytical Chemistry 2021, 140 , 116241. https://doi.org/10.1016/j.trac.2021.116241
- Yun-Yan Kuang, Xuan Gao, Yi-Jun Niu, Xun-Long Shi, Wei Zhou. Polymorphic Characterization, Pharmacokinetics, and Anti-Inflammatory Activity of Ginsenoside Compound K Polymorphs. Molecules 2021, 26
(7)
, 1983. https://doi.org/10.3390/molecules26071983
- Qonita Kurnia Anjani, Andi Dian Permana, Álvaro Cárcamo-Martínez, Juan Domínguez-Robles, Ismaiel A. Tekko, Eneko Larrañeta, Lalit K. Vora, Delly Ramadon, Ryan F. Donnelly. Versatility of hydrogel-forming microneedles in in vitro transdermal delivery of tuberculosis drugs. European Journal of Pharmaceutics and Biopharmaceutics 2021, 158 , 294-312. https://doi.org/10.1016/j.ejpb.2020.12.003
- Leena Peltonen, Clare J. Strachan. Degrees of order: A comparison of nanocrystal and amorphous solids for poorly soluble drugs. International Journal of Pharmaceutics 2020, 586 , 119492. https://doi.org/10.1016/j.ijpharm.2020.119492
- Beiqian Tian, Zhiyong Ding, Shuyi Zong, Jinyue Yang, Na Wang, Ting Wang, Xin Huang, Hongxun Hao. Manipulation of Pharmaceutical Polymorphic Transformation Process Using Excipients. Current Pharmaceutical Design 2020, 26
(21)
, 2553-2563. https://doi.org/10.2174/1381612826666200213122302
- Yijie Song, Yuanhua Cong, Bing Wang, Ning Zhang. Applications of Fourier transform infrared spectroscopy to pharmaceutical preparations. Expert Opinion on Drug Delivery 2020, 17
(4)
, 551-571. https://doi.org/10.1080/17425247.2020.1737671
- Marta F. Simões, Bernardo A. Nogueira, Andreia M. Tabanez, Rui Fausto, Rui M.A. Pinto, Sérgio Simões. Enhanced solid-state stability of amorphous ibrutinib formulations prepared by hot-melt extrusion. International Journal of Pharmaceutics 2020, 579 , 119156. https://doi.org/10.1016/j.ijpharm.2020.119156
- Joscha Brinkmann, Fabian Huxoll, Christian Luebbert, Gabriele Sadowski. Solubility of pharmaceutical ingredients in triglycerides. European Journal of Pharmaceutics and Biopharmaceutics 2019, 145 , 113-120. https://doi.org/10.1016/j.ejpb.2019.10.012
- Khadijah Edueng, Denny Mahlin, Johan Gråsjö, Olivia Nylander, Manish Thakrani, Christel A.S. Bergström. Supersaturation Potential of Amorphous Active Pharmaceutical Ingredients after Long-Term Storage. Molecules 2019, 24
(15)
, 2731. https://doi.org/10.3390/molecules24152731
- Emmi A.K. Palomäki, Jouko K. Yliruusi, Henrik V. Ehlers. Effect of headspace gas on nucleation of amorphous paracetamol. Journal of Drug Delivery Science and Technology 2019, 51 , 127-138. https://doi.org/10.1016/j.jddst.2019.02.019
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Dissolution profiles (drug release over time) for samples stored at (a) 30 °C/23% RH and (b) 30 °C/75% RH. Profiles of the pure amorphous tablet (day 0) and reference crystalline γ- and α-forms are shown for comparison. Profiles are generated with mean values of a minimum of three measurements. Error bars represent plus one standard deviation.
Figure 2
Figure 2. SEM micrographs of an amorphous indomethacin tablet (day 0, first column) and tablets stored for 1, 2, 7, and 22 days (from left to right) at 30 °C/23% RH before dissolution testing (top row). SEM images of the tablets prepared and stored in the same manner after 15 min of intrinsic dissolution testing (bottom row).
Figure 3
Figure 3. SEM micrographs of an amorphous indomethacin tablet (day 0, first column) and tablets stored for 1, 2, 7, and 22 days (from left to right) at 30 °C/75% RH before dissolution testing (top row). SEM images of the tablets prepared and stored in the same manner after 15 min of intrinsic dissolution testing (bottom row).
Figure 4
Figure 4. XRD diffractograms of tablets before and after 15 min of dissolution testing: (a) storage at 30 °C/23% RH and (b) storage at 30 °C/75% RH. Diffractograms of the α-, γ-, and ε-forms of indomethacin are shown for comparison, and some of their characteristic peaks are marked in green, red, and yellow, respectively. The ε-form was measured in transmission mode using a powder, while the α- and γ-forms were measured in reflection with tablets.
Figure 5
Figure 5. PCA scores plot of the ATR-FTIR spectra of the amorphous (day 0) and all stored samples after 15 min of dissolution testing (a); loadings and reference IR spectra of different solid-state forms of indomethacin (b).
Figure 6
Figure 6. CARS and SFG overlay images of samples stored at 30 °C/23% RH before (a) and after (b) 15 min of dissolution testing at pH 6.8. CARS spectra in the range of 1413–1800 cm–1 from selected regions marked by arrows plotted with reference spectra of amorphous, ε-, and γ-indomethacin (c–e). Overlay images (a, b) represent overlays of three channels: single CARS line at 1701 cm–1 in red (γ-indomethacin), single CARS line at 1676 cm–1 in blue (amorphous indomethacin), and a third channel representing the SFG signal (all noncentrosymmetric crystals) in green and yellow. The separation between green and yellow regions is based on the intensity ratios of CARS peaks at 1652 and 1676 cm–1 so that the regions having SFG activity and a CARS peak at 1652 cm–1 are colored green (α-indomethacin), and the regions having SFG activity and a CARS peak at 1676 cm–1 are colored yellow (ε-indomethacin). Panel (a) has been reprinted with permission from Novakovic et al. (18) Copyright 2017 American Chemical Society. am = amorphous.
Figure 7
Figure 7. CARS and SFG overlay images of samples stored at 30 °C/75% RH before (a) and after 15 min of dissolution testing at pH 6.8 (b). CARS spectra in the range of 1413–1800 cm–1 from selected regions marked by arrows (c–f) plotted with reference spectra of amorphous, ε-, α-, and γ-indomethacin. Overlay images (a, b) represent overlays of three channels: single CARS line at 1701 cm–1 in red (γ-indomethacin), single CARS line at 1676 cm–1 in blue (amorphous indomethacin), and a third channel representing the SFG signal (all noncentrosymmetric crystals) in green and yellow. The separation between green and yellow regions is based on the intensity ratios of CARS peaks at 1652 and 1676 cm–1 so that the regions having SFG activity and the CARS peak at 1652 cm–1 are colored green (α-indomethacin), and the regions having SFG activity and CARS peak at 1676 cm–1 are colored yellow (ε-indomethacin). Panel (a) has been reprinted with permission from Novakovic et al. (18) Copyright 2017 American Chemical Society. am = amorphous.
Figure 8
Figure 8. CARS (solid lines) and Raman (dotted lines) spectra of indomethacin solid-state forms. Raman spectra (except for the ε-form) are reproduced with permission from ref (23). The CARS spectral resolution is 12 cm–1, while the Raman spectral resolution is 4 cm–1. All CARS spectra are measured from the prepared reference forms, except for the spectrum of the η-form, which was recorded at the tablet surface. Polymorphs marked with asterisk are SFG-active.
References
This article references 47 other publications.
- 1European Directorate for the Quality of Medicines & HealthCare. European Pharmacopoeia 9.5. http://online6.edqm.eu/ep905/.There is no corresponding record for this reference.
- 2Hancock, B. C.; Parks, M. What is the true solubility advantage for amorphous pharmaceuticals?. Pharm. Res. 2000, 17 (4), 397– 404, DOI: 10.1023/A:10075167180482https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXktFalsb0%253D&md5=995cc8852439cc0319b3112a6abbc021What is the true solubility advantage for amorphous pharmaceuticals?Hancock, Bruno C.; Parks, MichaelPharmaceutical Research (2000), 17 (4), 397-404CODEN: PHREEB; ISSN:0724-8741. (Kluwer Academic/Plenum Publishers)In order to evaluate the magnitude of the soly. advantage for amorphous pharmaceutical materials when compared to their cryst. counterparts, the thermal properties of several drugs in their amorphous and cryst. states were detd. using differential scanning calorimetry. From these properties the soly. advantage for the amorphous form was predicted as a function of temp. using a simple thermodn. anal. These predictions were compared to the results of exptl. measurements of the aq. solubilities of the amorphous and cryst. forms of the drugs at several temps. By treating each amorphous drug as either an equil. supercooled liq. or a pseudo-equil. glass, the soly. advantage compared to the most stable cryst. form was predicted to be between 10 and 1600 fold. The measured soly. advantage was usually considerably less than this, and for one compd. studied in detail its temp. dependence was also less than predicted. It was calcd. that even for partially amorphous materials the apparent soly. enhancement (theor. or measured) is likely to influence in-vitro and in-vivo dissoln. behavior. Amorphous pharmaceuticals are markedly more sol. than their cryst. counterparts, however, their exptl. soly. advantage is typically less than that predicted from simple thermodn. considerations. This appears to be the result of difficulties in detg. the soly. of amorphous materials under true equil. conditions. Simple thermodn. predictions can provide a useful indication of the theor. max. soly. advantage for amorphous pharmaceuticals, which directly reflects the driving force for their initial dissoln.
- 3Murdande, S. B.; Pikal, M. J.; Shanker, R. M.; Bogner, R. H. Solubility advantage of amorphous pharmaceuticals: I. A thermodynamic analysis. J. Pharm. Sci. 2010, 99 (3), 1254– 1264, DOI: 10.1002/jps.219033https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmvVCqtg%253D%253D&md5=215a9f5a55ea378793b3eec9c263590aSolubility advantage of amorphous pharmaceuticals: I. A thermodynamic analysisMurdande, Sharad B.; Pikal, Michael J.; Shanker, Ravi M.; Bogner, Robin H.Journal of Pharmaceutical Sciences (2010), 99 (3), 1254-1264CODEN: JPMSAE; ISSN:0022-3549. (Wiley-Liss, Inc.)In recent years there has been growing interest in advancing amorphous pharmaceuticals as an approach for achieving adequate soly. Due to difficulties in the exptl. measurement of soly., a reliable est. of the soly. enhancement ratio of an amorphous form of a drug relative to its cryst. counterpart would be highly useful. We have developed a rigorous thermodn. approach to est. enhancement in soly. that can be achieved by conversion of a cryst. form to the amorphous form. We rigorously treat the three factors that contribute to differences in soly. between amorphous and cryst. forms. First, we calc. the free energy difference between amorphous and cryst. forms from thermal properties measured by modulated differential scanning calorimetry (MDSC). Secondly, since an amorphous solute can absorb significant amts. of water, which reduces its activity and soly., a correction is made using water sorption isotherm data and the Gibbs-Duhem equation. Next, a correction is made for differences in the degree of ionization due to differences in solubilities of the two forms. Utilizing this approach the theor. estd. soly. enhancement ratio of 7.0 for indomethacin (amorphous/γ-crystal) was found to be in close agreement with the exptl. detd. ratio of 4.9. © 2009 Wiley-Liss, Inc. and the American Pharmacists Assocn. J Pharm Sci 99: 1254-1264, 2010.
- 4Debnath, S.; Predecki, P.; Suryanarayanan, R. Use of glancing angle x-ray powder diffractometry to depth-profile phase transformations during dissolution of indomethacin and theophylline tablets. Pharm. Res. 2004, 21 (1), 149– 159, DOI: 10.1023/B:PHAM.0000012163.89163.f8There is no corresponding record for this reference.
- 5Ewing, A. V.; Clarke, G. S.; Kazarian, S. G. Stability of indomethacin with relevance to the release from amorphous solid dispersions studied with ATR-FTIR spectroscopic imaging. Eur. J. Pharm. Sci. 2014, 60, 64– 71, DOI: 10.1016/j.ejps.2014.05.0015https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXpvVagtbc%253D&md5=f0c5aea53b60d21b42d8da8076eebb6eStability of indomethacin with relevance to the release from amorphous solid dispersions studied with ATR-FTIR spectroscopic imagingEwing, Andrew V.; Clarke, Graham S.; Kazarian, Sergei G.European Journal of Pharmaceutical Sciences (2014), 60 (), 64-71CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)This work presents the use of attenuated total reflection Fourier transform IR (ATR-FTIR) spectroscopy and spectroscopic imaging to study the stability and dissoln. behavior of amorphous solid dispersions (ASDs). ASDs are employed to improve the bioavailability of drugs which are poorly sol. in aq. solns. Selecting the appropriate polymeric excipients for use in pharmaceutical tablets is crucial to control drug stability and subsequent release. In this study, indomethacin was used as a model poorly-aq. sol. drug since the amorphous-form has improved dissoln. properties over its cryst. forms. ASDs of indomethacin/polyethylene glycol (PEG) and indomethacin/hydroxypropyl methylcellulose (HPMC) in a 1:3 wt ratio were compared. Firstly, ATR-FTIR spectroscopy was employed to monitor the stability of indomethacin in the ASDs over 96 h. While the indomethacin/HPMC ASD showed the ability to maintain the amorphous indomethacin form for longer periods of time, ATR-FTIR spectra revealed that indomethacin in the drug/PEG ASD crystd. to the stable γ-form, via the α-form. Secondly, ATR-FTIR spectroscopic imaging was used to study the dissoln. of ASD tablets in a phosphate buffer (pH 7.5). Crystn. of amorphous indomethacin was characterized in the spectra collected during the dissoln. of the indomethacin/PEG ASD which consequently hindered release into the surrounding soln. In contrast, release of amorphous indomethacin was more effective from HPMC.
- 6Savolainen, M.; Kogermann, K.; Heinz, A.; Aaltonen, J.; Peltonen, L.; Strachan, C.; Yliruusi, J. Better understanding of dissolution behaviour of amorphous drugs by in situ solid-state analysis using Raman spectroscopy. Eur. J. Pharm. Biopharm. 2009, 71 (1), 71– 79, DOI: 10.1016/j.ejpb.2008.06.0016https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsV2lurfI&md5=4cbb70cb1088b3c72be715a8e848f0edBetter understanding of dissolution behaviour of amorphous drugs by in situ solid-state analysis using Raman spectroscopySavolainen, M.; Kogermann, K.; Heinz, A.; Aaltonen, J.; Peltonen, L.; Strachan, C.; Yliruusi, J.European Journal of Pharmaceutics and Biopharmaceutics (2009), 71 (1), 71-79CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)Amorphous drugs have a higher kinetic soly. and dissoln. rate than their cryst. counterparts. However, this advantage is lost if the amorphous form converts to the stable cryst. form during the dissoln. as the dissoln. rate will gradually change to that of the cryst. form. The purpose of this study was to use in situ Raman spectroscopy in combination with either partial least squares discriminant anal. (PLS-DA) or partial least squares (PLS) regression anal. to monitor as well as quantify the solid-phase transitions that take place during the dissoln. of two amorphous drugs, indomethacin (IMC) and carbamazepine (CBZ). The dissoln. rate was higher from amorphous IMC compared to the cryst. α- and γ-forms. However, the dissoln. rate started to slow down during the expt. In situ Raman anal. verified that at that time point the sample started to crystallize to the α-form. Amorphous CBZ instantly started to crystallize upon contact with the dissoln. medium. The transition from the amorphous form to CBZ dihydrate appears to go through the anhydrate form I. Based on the PLS anal. the amt. of form I formed in the sample during the dissoln. affected the dissoln. rate. Raman spectroscopy combined with PLS-DA was also more sensitive to the solid-state changes than X-ray powder diffraction (XRPD) and was able to detect changes in the solid-state that could not be detected with XRPD.
- 7Aaltonen, J.; Heinänen, P.; Peltonen, L.; Kortejärvi, H.; Tanninen, V. P.; Christiansen, L.; Hirvonen, J.; Yliruusi, J.; Rantanen, J. In situ measurement of solvent-mediated phase transformations during dissolution testing. J. Pharm. Sci. 2006, 95 (12), 2730– 2737, DOI: 10.1002/jps.20725There is no corresponding record for this reference.
- 8Greco, K.; Bogner, R. Crystallization of amorphous indomethacin during dissolution: Effect of processing and annealing. Mol. Pharmaceutics 2010, 7 (5), 1406– 1418, DOI: 10.1021/mp10001978https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmt1Kruro%253D&md5=4047d8e6882e5d4f5d9f001ce1f2c948Crystallization of Amorphous Indomethacin during Dissolution: Effect of Processing and AnnealingGreco, Kristyn; Bogner, RobinMolecular Pharmaceutics (2010), 7 (5), 1406-1418CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The crystn. of amorphous drugs during dissoln. is a type of soln. mediated phase transformation that can reduce the bioavailability enhancement one hoped to gain from the amorphous state. The goal of this study was to explore the effects of processing on the dissoln. performance of amorphous indomethacin. The amorphous solids were prepd. by four techniques, quench cooling the melted solid, cryogrinding γ indomethacin amorphous for 1 or 3 h and quench cooling the solid followed by 1 h of cryogrinding. Dissoln. results assessed in a flow-through intrinsic dissoln. app. reveal decreases in the dissoln. rate of amorphous indomethacin during the exptl. time frame indicating that a soln. mediated phase transformation has occurred. The amorphous solids prepd. by melt quenching and melt quenching followed by cryogrinding showed a significant dissoln. rate advantage over the γ form of indomethacin. In contrast, indomethacin that was cryoground amorphous for 1 or 3 h did not show any dissoln. rate advantage over the cryst. material. Transformation was confirmed by in situ Raman microscopy and polarized light microscopy with differences seen in the nature of the crystals apparent on the surface of the dissolving solid. A portion of the melt quenched amorphous sample was annealed at 25°C and 0% relative humidity to induce partial crystn. of γ indomethacin. As crystallinity increased, the dissoln. rate decreased. The transformation time of partially amorphous indomethacin was not dependent on the level of crystallinity present, indicating only a small fraction of cryst. material needs to be present to affect the kinetics of crystn. The soln. mediated phase transformation of amorphous indomethacin is affected by the processing method even though all solids were confirmed amorphous by polarized light microscopy and X-ray diffraction. Dissoln. may distinguish differences in amorphous solids that other methods cannot discern.
- 9Schmitt, P. D. Recent advances in nonlinear optical analyses of pharmaceutical materials in the solid state. Mol. Pharmaceutics 2017, 14 (3), 555– 565, DOI: 10.1021/acs.molpharmaceut.6b008099https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1Srsb8%253D&md5=55995e7bda16050a15aba0d39bc054c0Recent Advances in Nonlinear Optical Analyses of Pharmaceutical Materials in the Solid StateSchmitt, Paul D.Molecular Pharmaceutics (2017), 14 (3), 555-565CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)A review. The past decade has seen an increase in the use of nonlinear optical (NLO) techniques such as second harmonic generation, coherent antistokes Raman scattering, stimulated Raman scattering, and two-photon fluorescence for the solid-state characterization of pharmaceutical materials. These combined techniques offer several advantages (e.g., speed, selectivity, quantitation) of potential interest to the pharmaceutical community, as decreased characterization times in formulation development and testing could help decrease the time required to bring new, higher quality drugs to market. The large body of literature recently published in this field merits a review. Literature will be discussed in order of drug development, starting with applications in initial therapeutic mol. crystn. and polymorphic anal., followed by final dosage form characterization, and ending with drug product performance testing.
- 10Windbergs, M.; Jurna, M.; Offerhaus, H. L.; Herek, J. L.; Kleinebudde, P.; Strachan, C. J. Chemical imaging of oral solid dosage forms and changes upon dissolution using coherent anti-Stokes Raman scattering microscopy. Anal. Chem. 2009, 81 (6), 2085– 2091, DOI: 10.1021/ac802085610https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhslCisrc%253D&md5=4ecd9d725f53618dabc5d25210fc988aChemical imaging of oral solid dosage forms and changes upon dissolution using coherent anti-Stokes Raman scattering microscopyWindbergs, Maike; Jurna, Martin; Offerhaus, Herman L.; Herek, Jennifer L.; Kleinebudde, Peter; Strachan, Clare J.Analytical Chemistry (Washington, DC, United States) (2009), 81 (6), 2085-2091CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Dissoln. testing is a crucial part of pharmaceutical dosage form investigations and is generally performed by analyzing the concn. of the released drug in a defined vol. of flowing dissoln. medium. As solid-state properties of the components affect dissoln. behavior to a large and sometimes even unpredictable extent there is a strong need for monitoring and esp. visualizing solid-state properties during dissoln. testing. In this study coherent anti-Stokes Raman scattering (CARS) microscopy was used to visualize the solid-state properties of lipid-based oral dosage forms contg. the model drug theophylline anhydrate during dissoln. in real time. The drug release from the dosage form matrix was monitored with a spatial resoln. of about 1.5 μm. In addn., as theophylline anhydrate tends to form the less sol. monohydrate during dissoln., CARS microscopy allowed the solid-state transformation of the drug to be spatially visualized. The results obtained by CARS microscopy revealed that the method used to combine lipid and active ingredient into a sustained release dosage form can influence the physicochem. behavior of the drug during dissoln. In this case, formation of theophylline monohydrate on the surface was visualized during dissoln. with tablets compressed from powd. mixts. but not with solid lipid extrudates.
- 11Jurna, M.; Windbergs, M.; Strachan, C. J.; Hartsuiker, L.; Otto, C.; Kleinebudde, P.; Herek, J. L.; Offerhaus, H. L. Coherent anti-Stokes Raman scattering microscopy to monitor drug dissolution in different oral pharmaceutical tablets. J. Innovative Opt. Health Sci. 2009, 2 (1), 37– 43, DOI: 10.1142/S1793545809000322There is no corresponding record for this reference.
- 12Fussell, A.; Garbacik, E.; Offerhaus, H.; Kleinebudde, P.; Strachan, C. In situ dissolution analysis using coherent anti-Stokes Raman scattering (CARS) and hyperspectral CARS microscopy. Eur. J. Pharm. Biopharm. 2013, 85 (3, Part B), 1141– 1147, DOI: 10.1016/j.ejpb.2013.08.01212https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVylurrF&md5=e2c5dba6649cdfb115c1fd8c43765d3eIn situ dissolution analysis using coherent anti-Stokes Raman scattering (CARS) and hyperspectral CARS microscopyFussell, Andrew; Garbacik, Erik; Offerhaus, Herman; Kleinebudde, Peter; Strachan, ClareEuropean Journal of Pharmaceutics and Biopharmaceutics (2013), 85 (3PB), 1141-1147CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)The solid-state form of an active pharmaceutical ingredient (API) in an oral dosage form plays an important role in detg. the dissoln. rate of the API. As the solid-state form can change during dissoln., there is a need to monitor the oral dosage form during dissoln. testing. Coherent anti-Stokes Raman scattering (CARS) microscopy provides rapid, spectrally selective imaging to monitor the oral dosage form during dissoln. In this study, in situ CARS microscopy was combined with inline UV absorption spectroscopy to monitor the solid-state change in oral dosage forms contg. theophylline anhydrate undergoing dissoln. and to correlate the solid-state change with a change in dissoln. rate. The results from in situ CARS microscopy showed that theophylline anhydrate converted to theophylline monohydrate during dissoln. resulting in a redn. in the dissoln. rate. The addn. of Me cellulose to the dissoln. medium was found to delay the theophylline monohydrate growth and changed the morphol. of the monohydrate. The net effect was an increased dissoln. rate for theophylline anhydrate. Our results show that in situ CARS microscopy combined with inline UV absorption spectroscopy is capable of monitoring oral dosage forms undergoing dissoln. and correlating changes in solid-state form with changes in dissoln. rate.
- 13Kestur, U. S.; Wanapun, D.; Toth, S. J.; Wegiel, L. A.; Simpson, G. J.; Taylor, L. S. Nonlinear optical imaging for sensitive detection of crystals in bulk amorphous powders. J. Pharm. Sci. 2012, 101 (11), 4201– 4213, DOI: 10.1002/jps.2328013https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFWmtrnN&md5=8786157ff38e6777dea6847f3adf68feNonlinear optical imaging for sensitive detection of crystals in bulk amorphous powdersKestur, Umesh S.; Wanapun, Duangporn; Toth, Scott J.; Wegiel, Lindsay A.; Simpson, Garth J.; Taylor, Lynne S.Journal of Pharmaceutical Sciences (2012), 101 (11), 4201-4213CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)The primary aim of this study was to evaluate the utility of second-order nonlinear imaging of chiral crystals (SONICC) to quantify crystallinity in drug-polymer blends, including solid dispersions. Second harmonic generation (SHG) can potentially exhibit scaling with crystallinity between linear and quadratic depending on the nature of the source, and thus, it is important to det. the response of pharmaceutical powders. Phys. mixts. contg. different proportions of cryst. naproxen and hydroxyl Pr Me cellulose acetate succinate (HPMCAS) were prepd. by blending and a dispersion was produced by solvent evapn. A custom-built SONICC instrument was used to characterize the SHG intensity as a function of the cryst. drug fraction in the various samples. Powder X-ray diffraction (PXRD) and Raman spectroscopy were used as complementary methods known to exhibit linear scaling. SONICC was able to detect cryst. drug even in the presence of 99.9 wt % HPMCAS in the binary mixts. The calibration curve revealed a linear dynamic range with a R2 value of 0.99 spanning the range from 0.1 to 100 wt % naproxen with a root mean square error of prediction of 2.7%. Using the calibration curve, the errors in the validation samples were in the range of 5%-10%. Anal. of a 75 wt % HPMCAS-naproxen solid dispersion with SONICC revealed the presence of crystallites at an earlier time point than could be detected with PXRD and Raman spectroscopy. In addn., results from the crystn. kinetics expt. using SONICC were in good agreement with Raman spectroscopy and PXRD. In conclusion, SONICC has been found to be a sensitive technique for detecting low levels (0.1% or lower) of crystallinity, even in the presence of large quantities of a polymer. © 2012 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 14Wanapun, D.; Kestur, U. S.; Kissick, D. J.; Simpson, G. J.; Taylor, L. S. Selective detection and quantitation of organic molecule crystallization by second harmonic generation microscopy. Anal. Chem. 2010, 82 (13), 5425– 5432, DOI: 10.1021/ac100564f14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmvVSmsLo%253D&md5=1d176cb9495fe9eff0336a0faf80bd57Selective Detection and Quantitation of Organic Molecule Crystallization by Second Harmonic Generation MicroscopyWanapun, Duangporn; Kestur, Umesh S.; Kissick, David J.; Simpson, Garth J.; Taylor, Lynne S.Analytical Chemistry (Washington, DC, United States) (2010), 82 (13), 5425-5432CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Second order nonlinear optical imaging of chiral crystals (SONICC) was applied to selectively detect crystal formation at early stages and characterize the kinetics of nucleation and growth. SONICC relies on second harmonic generation (SHG), a nonlinear optical effect that only arises from noncentosym. ordered domain structures, which include crystals of chiral mols. The model systems studied include pharmaceutically relevant compds.: griseofulvin and chlorpropamide. SONICC demonstrates low detection limits producing an 8 order of magnitude improvement relative to macroscopic av. techniques and 5 order of magnitude improvement relative to optical microscopy. SONICC was also applied to examine the kinetics of crystn. in amorphous griseofulvin. The results show that SONICC enables simultaneous monitoring of individual crystal growth, nucleation rate, and macroscopic crystn. kinetics.
- 15Schmitt, P. D.; Trasi, N. S.; Taylor, L. S.; Simpson, G. J. Finding the needle in the haystack: Characterization of trace crystallinity in a commercial formulation of paclitaxel protein-bound particles by Raman spectroscopy enabled by second harmonic generation microscopy. Mol. Pharmaceutics 2015, 12 (7), 2378– 2383, DOI: 10.1021/acs.molpharmaceut.5b0006515https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVSht7nM&md5=244ef3d7eb97c0f6fdae03db367bb73dFinding the Needle in the Haystack: Characterization of Trace Crystallinity in a Commercial Formulation of Paclitaxel Protein-Bound Particles by Raman Spectroscopy Enabled by Second Harmonic Generation MicroscopySchmitt, Paul D.; Trasi, Niraj S.; Taylor, Lynne S.; Simpson, Garth J.Molecular Pharmaceutics (2015), 12 (7), 2378-2383CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Second harmonic generation (SHG) microscopy was used to rapidly identify regions of interest for localized confocal Raman spectroscopy measurements in order to quantify crystallinity within lyophilized Abraxane powder (protein bound paclitaxel for injectable suspension). Water insol. noncentrosym. cryst. particles ranging from ∼1 to 120 μm were identified by SHG, with wide variability in crystal size and frequency obsd. between several batches of Abraxane. By targeting the Raman anal. to these localized regions identified by SHG, the required measurement time was decreased over 2 orders of magnitude, from 8 h to 2 s. Exptl. Raman spectra of SHG active domains in Abraxane were in good agreement with exptl. spectra of pure cryst. paclitaxel. These collective results are consistent with up to 30% of the active ingredient being present as poorly sol. cryst. particulates in some batches of Abraxane.
- 16Mah, P. T.; Novakovic, D.; Saarinen, J.; Van Landeghem, S.; Peltonen, L.; Laaksonen, T.; Isomäki, A.; Strachan, C. J. Elucidation of compression-induced surface crystallization in amorphous tablets using sum frequency generation (SFG) microscopy. Pharm. Res. 2017, 34 (5), 957– 970, DOI: 10.1007/s11095-016-2046-6There is no corresponding record for this reference.
- 17Song, Z.; Sarkar, S.; Vogt, A. D.; Danzer, G. D.; Smith, C. J.; Gualtieri, E. J.; Simpson, G. J. Kinetic modeling of accelerated stability testing enabled by second harmonic generation microscopy. Anal. Chem. 2018, 90 (7), 4406– 4413, DOI: 10.1021/acs.analchem.7b0426017https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvFGns7w%253D&md5=a534778ce5b78e77924df5ba76416292Kinetic Modeling of Accelerated Stability Testing Enabled by Second Harmonic Generation MicroscopySong, Zhengtian; Sarkar, Sreya; Vogt, Andrew D.; Danzer, Gerald D.; Smith, Casey J.; Gualtieri, Ellen J.; Simpson, Garth J.Analytical Chemistry (Washington, DC, United States) (2018), 90 (7), 4406-4413CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)The low limits of detection afforded by second harmonic generation (SHG) microscopy coupled with image anal. algorithms enabled quant. modeling of the temp.-dependent crystn. of active pharmaceutical ingredients (APIs) within amorphous solid dispersions (ASDs). ASDs, in which an API is maintained in an amorphous state within a polymer matrix, are finding increasing use to address soly. limitations of small-mol. APIs. Extensive stability testing is typically performed for ASD characterization, the time frame for which is often dictated by the earliest detectable onset of crystal formation. Here a study of accelerated stability testing on ritonavir, a human immunodeficiency virus (HIV) protease inhibitor, has been conducted. Under the condition for accelerated stability testing at 50 °C/75%RH and 40 °C/75%RH, ritonavir crystn. kinetics from amorphous solid dispersions were monitored by SHG microscopy. SHG microscopy coupled by image anal. yielded limits of detection for ritonavir crystals as low as 10 ppm, which is about 2 orders of magnitude lower than other methods currently available for crystallinity detection in ASDs. The four decade dynamic range of SHG microscopy enabled quant. modeling with an established (JMAK) kinetic model. From the SHG images, nucleation and crystal growth rates were independently detd.
- 18Novakovic, D.; Saarinen, J.; Rojalin, T.; Antikainen, O.; Fraser-Miller, S. J.; Laaksonen, T.; Peltonen, L.; Isomäki, A.; Strachan, C. J. Multimodal nonlinear optical imaging for sensitive detection of multiple pharmaceutical solid-state forms and surface transformations. Anal. Chem. 2017, 89 (21), 11460– 11467, DOI: 10.1021/acs.analchem.7b0263918https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFOjt7rE&md5=02aa9d1d14293c87a2ac665153bfdde8Multimodal Nonlinear Optical Imaging for Sensitive Detection of Multiple Pharmaceutical Solid-State Forms and Surface TransformationsNovakovic, Dunja; Saarinen, Jukka; Rojalin, Tatu; Antikainen, Osmo; Fraser-Miller, Sara J.; Laaksonen, Timo; Peltonen, Leena; Isomaki, Antti; Strachan, Clare J.Analytical Chemistry (Washington, DC, United States) (2017), 89 (21), 11460-11467CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Two nonlinear imaging modalities, coherent anti-Stokes Raman scattering (CARS) and sum-frequency generation (SFG), were successfully combined for sensitive multimodal imaging of multiple solid-state forms and their changes on drug tablet surfaces. Two imaging approaches were used and compared: (i) hyperspectral CARS combined with principal component anal. (PCA) and SFG imaging and (ii) simultaneous narrowband CARS and SFG imaging. Three different solid-state forms of indomethacin-the cryst. gamma and alpha forms, as well as the amorphous form-were clearly distinguished using both approaches. Simultaneous narrowband CARS and SFG imaging was faster, but hyperspectral CARS and SFG imaging has the potential to be applied to a wider variety of more complex samples. These methodologies were further used to follow crystn. of indomethacin on tablet surfaces under two storage conditions: 30 °C/23% RH and 30 °C/75% RH. Imaging with (sub)micron resoln. showed that the approach allowed detection of very early stage surface crystn. The surfaces progressively crystd. to predominantly (but not exclusively) the gamma form at lower humidity and the alpha form at higher humidity. Overall, this study suggests that multimodal nonlinear imaging is a highly sensitive, solid-state (and chem.) specific, rapid, and versatile imaging technique for understanding and hence controlling (surface) solid-state forms and their complex changes in pharmaceuticals.
- 19Yamamoto, H. 1-Acyl-indoles. II. A new syntheses of 1-(p-chlorobenzoyl)-5-methoxy-3-indolylacetic acid and its polymorphism. Chem. Pharm. Bull. 1968, 16 (1), 17– 19, DOI: 10.1248/cpb.16.1719https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaF1cXkt1OitL0%253D&md5=3b9da89d4aaa5f1bd18f35caa43da1a21-Acyl-indoles. II. A new syntheses of 1-(ion-chlorobenzoyl)-5-methoxy-3-indolylacetic acid and its polymorphismYamamoto, HisaoChemical & Pharmaceutical Bulletin (1968), 16 (1), 17-19CODEN: CPBTAL; ISSN:0009-2363.1-(p-Chlorobenzoyl)-2-methyl-5-methoxy-3-indolylacetic acid, a potent antiinflammatory drug, was directly prepd. from N-(p-chlorobenzoyl)-p-methoxyphenyl-hydrazine-HCl and levulinic acid in excellent yield by the new method. The recrystn. of 1-(p-chlorobenzoyl)-2-methyl-5-methoxy-3-indolylacetic acid from solvents gave crystals of polymorphism -α, β, and γ-types.
- 20Borka, L. The Polymorphism of indomethacin. New modifications, their melting behavior and solubility. Acta Pharm. Suec. 1974, 11 (3), 295– 303There is no corresponding record for this reference.
- 21Lin, S. Y. Isolation and solid-state characteristics of a new crystal form of indomethacin. J. Pharm. Sci. 1992, 81 (6), 572– 576, DOI: 10.1002/jps.260081062221https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XkvFOiu7c%253D&md5=efd5a63b7b98cc610591f884fb4c662aIsolation and solid-state characteristics of a new crystal form of indomethacinLin, Shan YangJournal of Pharmaceutical Sciences (1992), 81 (6), 572-6CODEN: JPMSAE; ISSN:0022-3549.A new polymorphic crystal form of indomethacin was pptd. from an aq. soln. of indomethacin and β-cyclodextrin by titrn. with a 0.5N HCl aq. soln. Three polymorphs (α, β, and γ forms) and a new crystal form were differentiated with thermal anal., IR spectroscopy, powder x-ray diffractometry, TLC, elemental anal., and Fourier-transform IR (FTIR) spectroscopy with a newly developed FTIR microscope equipped with a thermal analyzer. The new crystal polymorph of indomethacin exhibited endo- and exothermic peaks near 102.8 and 104.1°, resp., because of phase transition without wt. loss, followed by 2 addnl. endothermic peaks at 151 and 158.9°, because of fusion. The differential scanning calorimetry-FTIR system can be used to examine the correlation between structural change of C:O stretching bands of this new polymorph and its thermal response. The new crystal form contained some γ-form crystals, detd. with an FTIR microscope equipped with a mapping option. Solid-state 13C-NMR spectra of the polymorphs of indomethacin were also examd.
- 22Crowley, K. J.; Zografi, G. Cryogenic grinding of indomethacin polymorphs and solvates: Assessment of amorphous phase formation and amorphous phase physical stability. J. Pharm. Sci. 2002, 91 (2), 492– 507, DOI: 10.1002/jps.1002822https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XhsVSms7g%253D&md5=c8cde5407ebb09c5062ae2debff18c39Cryogenic grinding of indomethacin polymorphs and solvates: assessment of amorphous phase formation and amorphous phase physical stabilityCrowley, Kieran J.; Zografi, GeorgeJournal of Pharmaceutical Sciences (2002), 91 (2), 492-507CODEN: JPMSAE; ISSN:0022-3549. (Wiley-Liss, Inc.)The effect of cryogenic grinding on five crystal forms of indomethacin (IMC) was investigated with particular interest in the formation of amorphous phase. Powder x-ray diffraction (PXRD) and differential scanning calorimetry (DSC) demonstrated that amorphous phase formation took place for all three polymorphs (γ, α, and δ) and one solvate (IMC methanolate). In the latter case, a postgrinding drying stage was needed to remove desolvated methanol from the ground amorphous product because methanol destabilized amorphous IMC presumably via a plasticizing effect. The crystal structure of another solvate, IMC t-butanolate, was unaffected by grinding, indicating that amorphous phase formation on grinding does not occur in all cases. Ground amorphous materials possessed similar glass transition temps. but significant differences in phys. stability as assessed by both isothermal and nonisothermal crystn. It is argued that phys. factors, namely residual crystal phase and sp. surface area, det. the isothermal and nonisothermal crystn. behavior of ground amorphous samples as opposed to intrinsic differences in the structure of the amorphous phase.
- 23Surwase, S. A.; Boetker, J. P.; Saville, D.; Boyd, B. J.; Gordon, K. C.; Peltonen, L.; Strachan, C. J. Indomethacin: New polymorphs of an old drug. Mol. Pharmaceutics 2013, 10 (12), 4472– 4480, DOI: 10.1021/mp400299a23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVWkur%252FI&md5=1d1974f7636c482223c74202c21af783Indomethacin: New Polymorphs of an Old DrugSurwase, Sachin A.; Boetker, Johan P.; Saville, Dorothy; Boyd, Ben J.; Gordon, Keith C.; Peltonen, Leena; Strachan, Clare J.Molecular Pharmaceutics (2013), 10 (12), 4472-4480CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)This study reports the appearance and characterization of multiple new polymorphic forms of indomethacin. Considering the interest in amorphous suspensions for toxicol. studies of poorly water-sol. drugs, we sought to investigate the crystn. behavior of amorphous indomethacin in aq. suspension. Specifically, the effect of pH and temp. on crystn. behavior was studied. Quench cooled amorphous powder was added to buffered media at different pH values (1.2, 4.5, and 6.8) at 5 and 25 °C. Both the solid and the soln. were analyzed at different time points up to 24 h. Attenuated total reflection Fourier transform IR (ATR-FTIR) spectroscopy (with principal component anal.) was used to study solid-phase transformations and UV spectroscopy used to probe soln. concn. The crystn. onset time decreased and rate of crystn. increased with increasing pH and temp. Diverse polymorphic forms were obsd., with three new forms being identified; these were named ε, ζ, and η. At 25 °C, the amorphous form recrystd. directly to the α form, except at pH 6.8, where it initially converted briefly into the ε form. At 5 °C, all three new polymorphic forms were obsd. sequentially in the order ε, ζ, and then η, with the no. of these forms obsd. increasing sequentially with decreasing pH. The three new forms exhibited distinct X-ray powder diffraction (XPRD), differential scanning calorimetry (DSC), and FTIR and Raman spectroscopy profiles. The soln. concn. profiles suggest that the relative phys. stabilities of the polymorphs at 5 °C from lowest to highest is ε < ζ < η < α. The appearance of new polymorphs in this study suggests that amorphous suspensions are worth considering when performing polymorphic screening studies.
- 24Van Duong, T.; Ludeker, D.; Van Bockstal, P. J.; De Beer, T.; Van Humbeeck, J.; Van den Mooter, G. Polymorphism of indomethacin in semicrystalline dispersions: Formation, transformation, and segregation. Mol. Pharmaceutics 2018, 15 (3), 1037– 1051, DOI: 10.1021/acs.molpharmaceut.7b0093024https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjs1agtQ%253D%253D&md5=3c27a34d72f18434d3e46ab6f38c4c0dPolymorphism of Indomethacin in Semicrystalline Dispersions: Formation, Transformation, and SegregationVan Duong, Tu; Ludeker, David; Van Bockstal, Pieter-Jan; De Beer, Thomas; Van Humbeeck, Jan; Van den Mooter, GuyMolecular Pharmaceutics (2018), 15 (3), 1037-1051CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The crystn. of metastable crystal polymorphs in polymer matrixes has been extensively reported in literature as a possible approach to enhance the soly. of poorly water-sol. drug compds., yet no clarification of the mechanism of the polymorph formation has been proposed. The current work aims to elucidate the polymorphism behavior of the model compd. indomethacin as well as the mechanism of polymorph selection of drugs in semicryst. systems. Indomethacin crystd. as either the α- or τ-form, a new metastable form, or a mixt. of the two polymorphs in dispersions contg. different drug loadings in polyethylene glycol, poloxamer, or Gelucire as the result of the variation in the mobility of drug mols. As a general rule, low mol. mobility of the amorphous drug favors the crystn. into thermodynamically stable forms whereas metastable cryst. polymorphs are preferred when the mol. mobility of the drug is sufficiently high. This rule provides insight into the polymorph selection of numerous active pharmaceutical ingredients in semicryst. dispersions and can be used as a guide for polymorphic screening from melt crystn. by tuning the mobility of drug mols. In addn., the drug crystd. faster while the polymer crystd. slower as the drug-loading increased with the maxima of drug crystn. rate in 70% indomethacin dispersion. Increasing the drug content in solid dispersions reduced the τ to α polymorphic transition rate, except for when the more stable form was initially dominant. The segregation of τ and α polymorphs as well as the polymorphic transformation during storage led to the inherent inhomogeneity of the semicryst. dispersions. This study highlights and expands our understanding about the complex crystn. behavior of semicryst. systems and is crucial for prepn. of solid dispersions with reproducible and consistent physicochem. properties and pharmaceutical performance.
- 25Kaneniwa, N.; Otsuka, M.; Hayashi, T. Physicochemical characterization of indomethacin polymorphs and the transformation kinetics in ethanol. Chem. Pharm. Bull. 1985, 33 (8), 3447– 3455, DOI: 10.1248/cpb.33.344725https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2MXmtVSrsrw%253D&md5=51415a5ce87b23c8d87e361fc1052889Physicochemical characterization of indomethacin polymorphs and the transformation kinetics in ethanolKaneniwa, Nobuyoshi; Otsuka, Makoto; Hayashi, TetsuoChemical & Pharmaceutical Bulletin (1985), 33 (8), 3447-55CODEN: CPBTAL; ISSN:0009-2363.Methods for the prepn. of polymorphs of indomethacin (IMC) (I) [53-86-1] were studied to obtain the pure polymorphs. The physicochem. properties of IMC polymorphs were measured by using x-ray diffraction anal. IR spectroscopy, DTA DSC, and 2 polymorphs (α and γ forms) and 1 benzene solvate (β form) were identified. The pure α form was obtained when distd. water at room temp. was poured into an EtOH [64-17-5] soln. of IMC at ∼80°, and the pptd. crystals were filtered and dried. The pure β and γ forms were obtained by recrystn. from benzene and Et2O, resp., at room temp. The m.ps. of the α and γ forms were 148 and 154°, resp., and their heats of fusion were 7.49 and 8.64 kcal/mol, resp., as detd. by DSC. A mixt. of α and γ forms was obtained by the method previously reported for α form prepn. (recrystn. method), since the pure α form was transformed to the γ form in EtOH at room temp. The transformation of α form to γ form in EtOH was analyzed by the kinetic method using 9 kinds of kinetic models. The transformation followed kinetics corresponding to 2-dimensional growth of nuclei (Avrami equation), and the activation energy was 14.2 kcal/mol from the Arrhenius plot. The solubilities of the α and γ forms in distd. water were 0.87 and 0.69 mg/100 mL, resp.
- 26Karmwar, P.; Graeser, K.; Gordon, K. C.; Strachan, C. J.; Rades, T. Effect of different preparation methods on the dissolution behaviour of amorphous indomethacin. Eur. J. Pharm. Biopharm. 2012, 80 (2), 459– 464, DOI: 10.1016/j.ejpb.2011.10.00626https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XitVCisLk%253D&md5=5dd34ccc6eeaea3d7228f6eb742d3ca4Effect of different preparation methods on the dissolution behaviour of amorphous indomethacinKarmwar, Pranav; Graeser, Kirsten; Gordon, Keith C.; Strachan, Clare J.; Rades, ThomasEuropean Journal of Pharmaceutics and Biopharmaceutics (2012), 80 (2), 459-464CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)The aim of this study was to investigate whether amorphous indomethacin samples prepd. using different preparative techniques and processing parameters exhibit different structural and thermodn. characteristics and whether these differences can be correlated to their dissoln. behavior. Samples were prepd. either by cooling the drug melt at different cooling rates or by cryo-milling the drug for different milling times. The resulting amorphous materials were characterized using X-ray diffraction, Raman spectroscopy and polarising light microscopy. All samples were entirely X-ray amorphous, except for the sample cryo-milled for 15 min, which exhibited residual crystallinity. The shape of the halos in the diffractograms, however, varied depending on the prepn. method and processing parameters, suggesting structural variations in the near order of the mols. between the prepd. amorphous forms. This finding was supported by principal component anal. of the Raman spectra, as the samples clustered in the scores plot according to processing parameters for both of the preparative methods used. When investigating the dissoln. behavior, the samples cooled at different cooling rates showed no significant differences in their dissoln. profiles and dissoln. rates (≈0.55 μg/mL/cm2). In contrast, for cryo-milled samples, dissoln. rate depended on the milling time, with samples milled for 120, 180 and 240 min, showing significantly increased dissoln. rates of 0.28, 0.48 and 0.59 μg/mL/cm2, resp., when compared to cryst. indomethacin (≈0.06 and 0.05 μg/mL/cm2 for α and γ-indomethacin, resp.). The milling processes appear to continue to affect the degree of disorder in the solid material, enhancing its dissoln. rate, although all samples milled for >30 min were X-ray amorphous. Thus, choosing the right prepn. technique and parameters for prepg. amorphous solids is crit. for producing materials with enhanced dissoln. profiles.
- 27Alonzo, D. E.; Zhang, G. G. Z.; Zhou, D.; Gao, Y.; Taylor, L. S. Understanding the behavior of amorphous pharmaceutical systems during dissolution. Pharm. Res. 2010, 27 (4), 608– 618, DOI: 10.1007/s11095-009-0021-127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhvV2gsLg%253D&md5=94caab8db7049285982b8a309dc579e9Understanding the Behavior of Amorphous Pharmaceutical Systems during DissolutionAlonzo, David E.; Zhang, Geoff G. Z.; Zhou, Deliang; Gao, Yi; Taylor, Lynne S.Pharmaceutical Research (2010), 27 (4), 608-618CODEN: PHREEB; ISSN:0724-8741. (Springer)Purpose: To investigate the underlying phys. processes taking place during dissoln. of amorphous pharmaceuticals and correlate them to the obsd. soln. concn.-time profiles. Felodipine and indomethacin were used as model hydrophobic compds. Concn.-time profiles were monitored during dissoln. of the model amorphous compds. using in situ fiber-optic UV spectroscopy. Crystn. of the solid exposed to an aq. environment was monitored using Raman spectroscopy and/or powder x-ray diffraction. Polarized light microscopy was used to provide qual. information about crystn. processes. For felodipine, a small extent of supersatn. was generated via dissoln. at 25°C but not at 37°C. The amorphous solid was found to crystallize rapidly at both temps. upon exposure to the dissoln. medium. Addn. of low concns. of polymers to the dissoln. medium was found to delay crystn. of the amorphous solid, leading to the generation of supersatd. solns. Amorphous indomethacin did not crystallize as readily in an aq. environment; hence, dissoln. resulted in supersatd. solns. However, crystn. from these supersatd. solns. was rapid. Polymeric additives were able to retard crystn. from supersatd. solns. of both indomethacin and felodipine for up to 4 h. The dissoln. advantage of amorphous solids can be negated either by crystn. of the amorphous solid on contact with the dissoln. medium or through rapid crystn. of the supersatd. soln. Polymeric additives can potentially retard both of these crystn. routes, leading to the generation of supersatd. solns. that can persist for biol. relevant timeframes.
- 28Mah, P. T.; Peltonen, L.; Novakovic, D.; Rades, T.; Strachan, C. J.; Laaksonen, T. The effect of surfactants on the dissolution behavior of amorphous formulations. Eur. J. Pharm. Biopharm. 2016, 103, 13– 22, DOI: 10.1016/j.ejpb.2016.03.00728https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XltFGqtLk%253D&md5=e196ac6a38a5877f7d914e4e15ea4757The effect of surfactants on the dissolution behavior of amorphous formulationsMah, Pei T.; Peltonen, Leena; Novakovic, Dunja; Rades, Thomas; Strachan, Clare J.; Laaksonen, TimoEuropean Journal of Pharmaceutics and Biopharmaceutics (2016), 103 (), 13-22CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)The optimal design of oral amorphous formulations benefits from the use of excipients to maintain drug supersatn. and thus ensures adequate absorption during intestinal transit. The use of surfactants for the maintenance of supersatn. in amorphous formulations has not been investigated in detail. The main aim of this study was to investigate the effect of surfactant on the dissoln. behavior of neat amorphous drug and binary polymer based solid dispersion. Indomethacin was used as the model drug and the surfactants studied were polysorbate 80 and poloxamer 407. The presence of surfactants (alone or in combination with polymers) in the buffer was detrimental to the dissoln. of neat amorphous indomethacin, suggesting that the surfactants promoted the crystn. of neat amorphous indomethacin. In contrast, the presence of surfactants (0.01% w/v) in the buffer resulted in a significant improvement on the dissoln. behavior of binary polymer based solid dispersion. Incorporating the surfactant to the formulation to form ternary solid dispersion adversely affected the dissoln. behavior. In conclusion, the use of surfactants (as wetting or solubilization agents) in dissoln. studies of neat amorphous drugs requires prudent consideration. The design of amorphous formulations with optimal dissoln. performance requires the appropriate selection of a combination of excipients and consideration of the method of introducing the excipients.
- 29Karmwar, P.; Graeser, K.; Gordon, K. C.; Strachan, C. J.; Rades, T. Investigation of properties and recrystallisation behaviour of amorphous indomethacin samples prepared by different methods. Int. J. Pharm. 2011, 417 (1–2), 94– 100, DOI: 10.1016/j.ijpharm.2010.12.01929https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVygsL3M&md5=c6177699d554012c2923f9d1d868e4d0Investigation of properties and recrystallisation behaviour of amorphous indomethacin samples prepared by different methodsKarmwar, Pranav; Graeser, Kirsten; Gordon, Keith C.; Strachan, Clare J.; Rades, ThomasInternational Journal of Pharmaceutics (2011), 417 (1-2), 94-100CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)The aim of this study was to investigate if amorphous indomethacin samples, prepd. using different prepn. methods, exhibit different structural and kinetic characteristics and if these differences can be correlated to their phys. stability (time to crystn.). Samples were prepd. by melt quenching, spray drying, ball milling, and cryo-milling. The resulting amorphous materials were characterized using X-ray diffraction, Raman spectroscopy and differential scanning calorimetry. All freshly prepd. samples were completely X-ray amorphous (with a halo being the only feature in the diffractograms). The shape of the halos in the diffractograms, however, varied depending on the prepn. method, suggesting structural variations in the near order of the mols. between the differently prepd. amorphous forms. Principal component anal. of the Raman spectra of the various amorphous forms revealed that the samples clustered in the scores plot according to prepn. method, again suggesting structural differences due to prepn. method. The range of vibrations assocd. with the largest spectral differences in the loadings plot showed that these differences were due to a range of mol. conformations and intermol. interactions. The ranking of the samples with respect to stability was: quench cooled amorphous samples > cryo-milled (α-form) > spray dried > ball milled (α-form) > ball milled (γ-form) = cryo-milled (γ-form). This ranking was not correlated with the diffractogram shapes or sample distribution in the scores plot of the Raman spectra, suggesting that phys. stability was not directly affected by structural variation in the samples. However, ranking of stability of the differently prepd. amorphous forms of the drug could be predicted by detg. the relaxation time values, for all amorphous samples. The relaxation times, calcd. by using the Adam Gibbs and Kohlrausch-Williams-Watts equations, were in accordance with the exptl. detd. stability order. This study showed that correlation of phys. stability with calcd. relaxation time is possible for the same amorphous systems prepd. by different methods. This could aid in selecting the most appropriate prepn. techniques in situations where there are a variety of suitable methods.
- 30Wu, T.; Yu, L. Surface crystallization of indomethacin below Tg. Pharm. Res. 2006, 23 (10), 2350– 2355, DOI: 10.1007/s11095-006-9023-430https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtVeqsbrF&md5=2c96d63050cd5154235bc4ecea6a3949Surface Crystallization of Indomethacin Below Glass Transition TemperatureWu, Tian; Yu, LianPharmaceutical Research (2006), 23 (10), 2350-2355CODEN: PHREEB; ISSN:0724-8741. (Springer)Purpose. To study the surface crystn. of indomethacin (IMC) below T g and its effects on the kinetics of overall crystn. Methods. Crystal growth rates in liq. layers formed between microscope cover glasses were measured with the top cover glass in place and removed. Polymorphs were identified by powder x-ray diffraction, Raman microscopy, and melting-point detn. by hot-stage microscopy. Surface crystals were identified by scratching the sample surface, by cutting the sample to expose its interior, and by analyzing the intensity of x-ray diffraction. Amorphous IMC particles of different sizes were stored at 40° (T g-2°) and analyzed at different times by differential scanning calorimetry to obtain the kinetics of crystn. Results. Crystal growth of IMC below T g at the free surface was approx. two orders of magnitude faster than that in the bulk, resulting in a surface layer of crystals around a slower-crystg. interior. Surface crystn. yielded mainly the γ polymorph. Amorphous IMC powders showed rapid initial crystn. at 40°, but the crystn. abruptly slowed down at "satn. levels" below 100%; the larger the particles, the lower the "satn. level." Conclusion. The faster surface crystn. of IMC than the bulk crystn. leads to unusual crystn. kinetics wherein a rapid initial increase of crystallinity is followed by an abrupt slowdown of crystn. Surface crystn. should be distinguished from bulk crystn. in modeling and controlling the crystn. of amorphous solids.
- 31Andronis, V.; Yoshioka, M.; Zografi, G. Effects of sorbed water on the crystallization of indomethacin from the amorphous state. J. Pharm. Sci. 1997, 86 (3), 346– 351, DOI: 10.1021/js960271131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXhtVGhtrs%253D&md5=c5bdb49f1308a0812dc207822279dd12Effects of Sorbed Water on the Crystallization of Indomethacin from the Amorphous StateAndronis, Vlassios; Yoshioka, Minoru; Zografi, GeorgeJournal of Pharmaceutical Sciences (1997), 86 (3), 346-351CODEN: JPMSAE; ISSN:0022-3549. (American Chemical Society)The water sorption isotherm and the crystn. rates for amorphous indomethacin were detd. at 30° as a function of relative humidity (RH), along with the effects of water content on the glass transition temp. (Tg). Below 43% RH, only the stable γ crystal form appears, whereas at higher RH, only the metastable α crystal form appears. The tendency for the α crystals to form at higher RH is consistent with the Ostwald step rule. The crystn. rate of the α form continuously increased with increasing RH due to increasing mol. mobility. The crystn. mechanism of the γ form changed from surface-initiated to bulk-initiated crystn. at 21% RH, and although crystn. rates of the γ form increased with increasing RH in both cases, they were higher when crystn. was surface-initiated. The complex crystn. behavior of the γ form is explained by the higher water content and mol. mobility of the surface relative to the bulk and the general effect of water on α or γ crystal form selection as described by the Ostwald step rule.
- 32Priemel, P. A.; Grohganz, H.; Gordon, K. C.; Rades, T.; Strachan, C. J. The impact of surface- and nano-crystallisation on the detected amorphous content and the dissolution behaviour of amorphous indomethacin. Eur. J. Pharm. Biopharm. 2012, 82 (1), 187– 193, DOI: 10.1016/j.ejpb.2012.05.01232https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtV2mtLnL&md5=9c9d7f4b2578d1d064d66cf4691b0945The impact of surface- and nano-crystallisation on the detected amorphous content and the dissolution behaviour of amorphous indomethacinPriemel, P. A.; Grohganz, H.; Gordon, K. C.; Rades, T.; Strachan, C. J.European Journal of Pharmaceutics and Biopharmaceutics (2012), 82 (1), 187-193CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)The crystallinity and phys. stability of amorphous drugs has previously been studied using different anal. techniques. However, the effect of the measurement method on obsd. crystallinity and its importance for crit. quality attributes, such as dissoln., has not yet been widely investigated.The aim of this study was to (i) qual. analyze and understand the recrystn. behavior of amorphous indomethacin during storage, (ii) quantify the amorphous content during storage with complementary anal. techniques and (iii) investigate the relationship between obsd. recrystn. behavior and dissoln. behavior.Quench cooled indomethacin was stored and the samples were visualized by SEM to gain spatially resolved information about the recrystn. behavior. Crystn. was quantified by Fourier transform attenuated total reflectance IR (FT-ATR-IR) spectroscopy, differential scanning calorimetry and X-ray powder diffraction.These techniques resulted in different obsd. recrystn. profiles. The physicochem. phenomena detected and sampling geometry for each technique together with the sample recrystg. from the surface and appearance of nano-crystals were used to explain the differences.The dissoln. behavior at the obsd. recrystn. endpoints for the different anal. techniques revealed that FT-ATR-IR spectroscopy predicted the changes in dissoln. behavior due to crystn. best.
- 33Tres, F.; Treacher, K.; Booth, J.; Hughes, L. P.; Wren, S. A. C.; Aylott, J. W.; Burley, J. C. Indomethacin-Kollidon VA64 extrudates: A mechanistic study of pH-dependent controlled release. Mol. Pharmaceutics 2016, 13 (3), 1166– 1175, DOI: 10.1021/acs.molpharmaceut.5b0097933https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvV2rs7k%253D&md5=9cbf9d0af9b46777e849b7ccc6e8b36cIndomethacin-Kollidon VA64 Extrudates: A Mechanistic Study of pH-Dependent Controlled ReleaseTres, Francesco; Treacher, Kevin; Booth, Jonathan; Hughes, Leslie P.; Wren, Stephen A. C.; Aylott, Jonathan W.; Burley, Jonathan C.Molecular Pharmaceutics (2016), 13 (3), 1166-1175CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Because of its weakly acidic nature (pKa of 4.5), indomethacin presents an aq. soly. that significantly increases when changing from acidic to neutral/alk. pH (1.5 μg/mL at pH 1.2 and 105.2 μg/mL at pH 7.4). We have therefore investigated the impact of the dissoln. medium pH on the dissoln. performance of indomethacin:Kollidon VA64 extrudates. The impact of the drug loading on the dissoln. properties of these systems was also examd. (5%, 15%, 30%, 50%, 70%, and 90% drug loading). Time-resolved Raman spectroscopy along with in-line UV-vis spectrophotometry was employed to directly relate changes in dissoln. behavior to physicochem. changes that occur to the extrudate during the test. The dissoln. tests were performed in pH 2 HCl (to mimic the stomach conditions), and this was then switched during the expt. to pH 6.8 phosphate buffer (to simulate the poststomach conditions). The rotating disk dissoln. rate test was also used to simultaneously measure the dissoln. rate of both the drug and the polymer. We found that in pH 2 HCl buffer, for the 15% or higher drug-loaded extrudates, Kollidon VA64 preferentially dissolves from the exterior of the compact leaving an amorphous drug-rich hydrophobic shell, which, similarly to an enteric coating, inhibits the drug release. The in situ formation of an enteric coating has been previously hypothesized, and this has been the first time that is directly obsd. in a pH-variable dissoln. test. The dissoln. medium switch to pH 6.8 phosphate buffer, due to the large increase of the aq. soly. of indomethacin at this pH, leads to rapid dissoln. of the material forming the coating and therefore total drug release. In contrast, the 5% extrudate is fully hydrated and quickly dissolves at low pH pointing to a dissoln. performance dependent on highly water-sol. Kollidon VA64.
- 34Koranne, S.; Thakral, S.; Suryanarayanan, R. Effect of formulation and process parameters on the disproportionation of indomethacin sodium in buffered lyophilized formulations. Pharm. Res. 2018, 35 (11), 214, DOI: 10.1007/s11095-018-2499-xThere is no corresponding record for this reference.
- 35Peltonen, L.; Liljeroth, P.; Heikkilä, T.; Kontturi, K.; Hirvonen, J. Dissolution testing of acetylsalicylic acid by a channel flow method—correlation to USP basket and intrinsic dissolution methods. Eur. J. Pharm. Sci. 2003, 19 (5), 395– 401, DOI: 10.1016/S0928-0987(03)00140-435https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXlvFyiurY%253D&md5=6df5aa626ee4587f80bd35328bb4bf29Dissolution testing of acetylsalicylic acid by a channel flow method-correlation to USP basket and intrinsic dissolution methodsPeltonen, Leena; Liljeroth, Peter; Heikkila, Tiina; Kontturi, Kyosti; Hirvonen, JouniEuropean Journal of Pharmaceutical Sciences (2003), 19 (5), 395-401CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)A new modification of the channel flow dissoln. method is introduced together with the theor. basis to ext. the soly. and mass transfer parameters from the dissoln. expts. Correlation of drug dissoln. profiles in the channel flow app. was evaluated with respect to USP basket and intrinsic dissoln. methods at pH 1.2 or 6.8. Acetylsalicylic acid (ASA) was studied as a pure drug substance and as three simple tablet compns. with microcryst. cellulose (MCC) and/or lactose as excipients. The channel flow measurements of 100% ASA tablets correlated well with the results of intrinsic dissoln. tests. In the channel flow method as well as in the USP basket method the release of ASA was fastest from the tablet compns. contg. lactose, while the slowest dissoln. rate was obsd. with the compn. contg. MCC as the only excipient. As presumed, the dissoln. rate of the weak acid was decreased as the pH of the medium was lowered, which was clearly confirmed also by the three dissoln. methods. MCC forms matrix tablets and in the USP basket method the dissoln. profiles followed square root of time kinetics indicating that diffusion was the rate-controlling step of ASA dissoln. Also the channel flow results indicated that the dissoln. Of ASA was controlled by mass transfer. The swelling behavior of the tablets is different in the channel flow method as compared to the basket method: only one tablet surface is exposed to the dissoln. medium in the channel flow system. The contact between the tablet surface and the dissoln. medium is more similar between the channel flow and intrinsic dissoln. methods.
- 36Sarnes, A.; Østergaard, J.; Jensen, S. S.; Aaltonen, J.; Rantanen, J.; Hirvonen, J.; Peltonen, L. Dissolution study of nanocrystal powders of a poorly soluble drug by UV imaging and channel flow methods. Eur. J. Pharm. Sci. 2013, 50 (3), 511– 519, DOI: 10.1016/j.ejps.2013.08.03036https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFCitLvK&md5=ef415dcf6bbdd663d96b5e9d9693813aDissolution study of nanocrystal powders of a poorly soluble drug by UV imaging and channel flow methodsSarnes, Annika; Oestergaard, Jesper; Jensen, Sabrine Smedegaard; Aaltonen, Jaakko; Rantanen, Jukka; Hirvonen, Jouni; Peltonen, LeenaEuropean Journal of Pharmaceutical Sciences (2013), 50 (3-4), 511-519CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)Application of drug nanocrystals provides advantageous options for the pharmaceutical formulation development of poorly sol. drugs. The objective of this study was to investigate the dissoln. behavior improving effects of differently sized nanocrystals of a poorly sol. model drug, indomethacin. Nanocrystal suspensions were prepd. using a top-down wet milling technique with three stabilizers: poloxamer F68, poloxamer F127 and polysorbate 80. The dissoln. of the differently sized indomethacin nanocrystals were investigated using a channel flow dissoln. method and by UV imaging. Unmilled bulk indomethacin and phys. mixts. were used as refs. According to both the dissoln. methods, the dissoln. properties of indomethacin were improved by the particle size redn. UV imaging was used for the first time as a dissoln. testing method for fast dissolving nanoscale material. The technique provided new information about the concn. of the dissolved drug next to the sample surface; with the smallest nanocrystals (580 nm) the indomethacin concn. next to the particle surface exceeded five-fold the thermodn. satd. indomethacin soln. concn. Thus the soly. improvement itself, not only the increased surface area for dissoln., may have an important role in the higher dissoln. rates of nanocrystal formulations. Poloxamer F68 was the most optimal stabilizer in the prepn. of the indomethacin nanocrystal suspensions and in the soly. and dissoln. enhancement as well.
- 37Sunesen, V. H.; Pedersen, B. L.; Kristensen, H. G.; Müllertz, A. In vivo in vitro correlations for a poorly soluble drug, danazol, using the flow-through dissolution method with biorelevant dissolution media. Eur. J. Pharm. Sci. 2005, 24 (4), 305– 313, DOI: 10.1016/j.ejps.2004.11.00737https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhslWmsLc%253D&md5=e3fca526cc00dffbeadd9b1c856eb658In vivo in vitro correlations for a poorly soluble drug, danazol, using the flow-through dissolution method with biorelevant dissolution mediaSunesen, Vibeke Hougaard; Pedersen, Betty Lomstein; Kristensen, Henning Gjelstrup; Muellertz, AnetteEuropean Journal of Pharmaceutical Sciences (2005), 24 (4), 305-313CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)The purpose of the study was to design dissoln. tests that were able to distinguish between the behavior of danazol under fasted and fed conditions, by biorelevant media. In vitro dissoln. of 100 mg danazol capsules was performed using the flow-through dissoln. method. Flow rates were 8, 16 or 32 mL/min, corresponding to total vols. dissoln. medium of 960, 1920 and 3840 mL, resp. The media used contained bile salt and phospholipid levels relevant for either fasted or fed conditions in vivo. Crude and inexpensive bile components, Porcine Bile Ext. and soybean phospholipids, were used as the bile source. The effect of adding different concns. and molar ratios of monoglycerides and fatty acids to the fed state media was investigated. In vivo release profiles under fasted and fed conditions were obtained from a previous study by deconvolution [Sunesen, V.H., Vedelsdal, R., Kristensen, H.G., Christrup, L., Muellertz, A. 2005]. Effect of liq. vol. and food intake on the abs. bioavailability of danazol, a poorly sol. drug, [Eur. J. Pharm. Sci. 24, 297-303]. In the fasted state, the physiol. most relevant correlation with in vivo results was achieved with a medium contg. 6.3 mM bile salts and 1.25 mM phospholipids (8 mL/min). A medium contg. 18.8 mM bile salts, 3.75 mM phospholipids, 4.0 mM monoglycerides and 30 mM fatty acids (8 mL/min) gave the closest correlation with fed state in vivo results. By using the flow-through dissoln. method it was possible to obtain correlations with in vivo release of danazol under fasted and fed conditions. Both hydrodynamics and medium compn. were important for the dissoln. of danazol. In the fed state an IVIVC could only be obtained by including monoglycerides and fatty acids in the medium.
- 38Greco, K.; Bogner, R. Solution-mediated phase transformation: Significance during dissolution and implications for bioavailability. J. Pharm. Sci. 2012, 101 (9), 2996– 3018, DOI: 10.1002/jps.2302538https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1OltbnF&md5=f94251f8c92af8013a346b316823bf4fSolution-mediated phase transformation: Significance during dissolution and implications for bioavailabilityGreco, Kristyn; Bogner, RobinJournal of Pharmaceutical Sciences (2012), 101 (9), 2996-3018CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)A review. Soly. improvement of poorly sol. drug compds. is a key approach to ensuring the successful development of many new drugs. Methods used to improve the soly. of drug compds. include forming a salt, cocrystal, or amorphous solid. These methods of improving soly. can often lead to a phenomenon called soln.-mediated phase transformation, a phase change that is facilitated through exposure to soln. Soln.-mediated phase transformation occurs in three steps: dissoln. to create a supersatd. soln. followed by nucleation of less sol. phase and the growth of that phase. When the growth of the less sol. phase occurs on the surface of the metastable solid, this phenomenon can cause a marked decrease in dissoln. rate during in vitro dissoln. evaluation, and ultimately in vivo. Therefore, transformation to a less sol. solid during dissoln. is an important aspect to consider when evaluating approaches to increase the soly. of a poorly sol. drug. Identification of soln.-mediated phase transformation during dissoln. is reviewed for powder dissoln., rotating disk method, and channel flow-through app. Types of soln.-mediated phase transformation are described in this report, including those involving salts, polymorphs, amorphous solids, and cocrystals. Many exptl. examples are provided. Evidence of potential soln.-mediated phase transformation in vivo is discussed to better understand the relationship between in vitro dissoln. evaluation and in vivo performance. © 2011 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 39Slavin, P. A.; Sheen, D. B.; Shepherd, E. E. A.; Sherwood, J. N.; Feeder, N.; Docherty, R.; Milojevic, S. Morphological evaluation of the γ-polymorph of indomethacin. J. Cryst. Growth 2002, 237–239, 300– 305, DOI: 10.1016/S0022-0248(01)01924-839https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XjvVSktr8%253D&md5=ecfc82d311655b5cc96384253b4eef3aMorphological evaluation of the γ-polymorph of indomethacinSlavin, Paul A.; Sheen, David B.; Shepherd, Evelyn E. A.; Sherwood, John N.; Feeder, Neil; Docherty, Robert; Milojevic, SnezenaJournal of Crystal Growth (2002), 237-239 (Pt. 1), 300-305CODEN: JCRGAE; ISSN:0022-0248. (Elsevier Science B.V.)A study was made of the polymorphic nature of crystals of the pharmaceutical indomethacin grown from a wide range of solvents and from the melt. In most solvents, growth at high supersaturations yielded either a 1:0.5 solvated form (approx.) or the α-polymorph. At low supersaturations the γ-polymorph was commonly produced. Solns. in MeOH and tBuOH yielded a 1:1 solvate. The morphol. of the γ-form showed no variation with solvent type but changed with supersatn. in a manner consistent with a differential variation in growth rates of the faces. This lack of solvent influence was confirmed by the fact that a similar morphol. resulted on growth from the melt. Morphol. predictions were carried out for the γ-polymorph and these show good agreement with exptl. observations.
- 40Hartshorn, C. M.; Lee, Y. J.; Camp, C. H.; Liu, Z.; Heddleston, J.; Canfield, N.; Rhodes, T. A.; Hight Walker, A. R.; Marsac, P. J.; Cicerone, M. T. Multicomponent chemical imaging of pharmaceutical solid dosage forms with broadband CARS microscopy. Anal. Chem. 2013, 85 (17), 8102– 8111, DOI: 10.1021/ac400671pThere is no corresponding record for this reference.
- 41Taylor, L. S.; Zografi, G. Spectroscopic characterization of interactions between PVP and indomethacin in amorphous molecular dispersions. Pharm. Res. 1997, 14 (12), 1691– 1698, DOI: 10.1023/A:101216741037641https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXktFKjtQ%253D%253D&md5=9bc968c8777589f6d0d90b2f349237ddSpectroscopic characterization of interactions between PVP and indomethacin in amorphous molecular dispersionsTaylor, Lynne S.; Zografi, GeorgePharmaceutical Research (1997), 14 (12), 1691-1698CODEN: PHREEB; ISSN:0724-8741. (Plenum Publishing Corp.)The mol. structure of indomethacin-PVP amorphous solid dispersions was studied and specific interactions between the components was identified using vibrational spectroscopy. Solid dispersions of PVP and indomethacin were prepd. using a solvent evapn. technique and IR and FT-Raman spectra were obtained. A comparison of the carbonyl stretching region of γ indomethacin, known to form carboxylic acid dimers, with that of amorphous indomethacin indicated that the amorphous phase exists predominantly as dimers. The hydrogen bonding of α indomethacin is not as dimers. Addn. of PVP to amorphous indomethacin increased the intensity of the IR band assigned to non-hydrogen bonded carbonyl. Concomitantly, the PVP carbonyl stretch appeared at a lower wavenumber indicating hydrogen bonding. Model solvent systems aided spectral interpretation. The magnitude of the spectral changes were comparable for an indomethacin-PVP solid dispersion and a soln. of indomethacin in methylpyrrolidone at the same wt. percent. Indomethacin interacts with PVP in solid dispersions through hydrogen bonds formed between the drug hydroxyl and polymer carbonyl resulting in disruption of indomethacin dimers. PVP may influence the crystn. kinetics by preventing the self assocn. of indomethacin mols. The similarity of results for solid dispersions and solns. emphasizes the "soln." nature of this binary amorphous state.
- 42Trasi, N. S.; Purohit, H. S.; Taylor, L. S. Evaluation of the crystallization tendency of commercially available amorphous tacrolimus formulations exposed to different stress conditions. Pharm. Res. 2017, 34 (10), 2142– 2155, DOI: 10.1007/s11095-017-2221-442https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFCrsbvP&md5=4d1019be54fe12ef75f30251a14f6869Evaluation of the Crystallization Tendency of Commercially Available Amorphous Tacrolimus Formulations Exposed to Different Stress ConditionsTrasi, Niraj S.; Purohit, Hitesh S.; Taylor, Lynne S.Pharmaceutical Research (2017), 34 (10), 2142-2155CODEN: PHREEB; ISSN:0724-8741. (Springer)Tacrolimus, an immunosuppressant, is a poorly water sol. compd. whereby the com. available capsule formulations contain the drug in amorphous form. The goal of this study was to evaluate the robustness of the innovator product and five generic formulations to crystn. following storage at stress conditions. Methods: Products were purchased from a pharmacy and stored at 40/75% relative humidity (RH), open dish conditions. Crystallinity was detd. using x-ray diffraction. The quantity of the ingredients in the formulations were detd. using different approaches and the various factors that might cause instability in the formulations were studied. Results: After 4 wk of open dish storage at 40/75% RH, one of the generic formulations showed evidence of tacrolimus crystn. Further investigations revealed batch-to-batch variations in crystn. tendency with the extent of crystallinity varying between 50 and 100% for different batches. Crystn. was also obsd. at lower storage temps. (30) when the RH was maintained at 75%. It was found that crystn. could be induced in a model formulation by wet granulating an ethanolic soln. of the drug with lactose and drying at 60-70 followed by exposure to stress conditions. Conclusions: It seems probable that the generic that was susceptible to crystn. contains amorphous drug phys. mixed with polymeric excipients, rather than as an amorphous solid dispersion. This study highlights the importance of considering the manufg. process on the stability of the resultant amorphous product.
- 43Sun, D. D.; Lee, P. I. Evolution of supersaturation of amorphous pharmaceuticals: The effect of rate of supersaturation generation. Mol. Pharmaceutics 2013, 10 (11), 4330– 4346, DOI: 10.1021/mp400439q43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFSms7%252FO&md5=d7452e8a147c4f673223032871751f51Evolution of Supersaturation of Amorphous Pharmaceuticals: The Effect of Rate of Supersaturation GenerationSun, Dajun D.; Lee, Ping I.Molecular Pharmaceutics (2013), 10 (11), 4330-4346CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The combination of a rapidly dissolving and supersaturating spring with a pptn. retarding parachute has often been pursued as an effective formulation strategy for amorphous solid dispersions (ASDs) to enhance the rate and extent of oral absorption. However, the interplay between these two rate processes in achieving and maintaining supersatn. remains inadequately understood, and the effect of rate of supersatn. buildup on the overall time evolution of supersatn. during the dissoln. of amorphous solids has not been explored. The objective of this study is to investigate the effect of supersatn. generation rate on the resulting kinetic soly. profiles of amorphous pharmaceuticals and to delineate the evolution of supersatn. from a mechanistic viewpoint. Exptl. concn.-time curves under varying rates of supersatn. generation and recrystn. for model drugs, indomethacin (IND), naproxen (NAP) and piroxicam (PIR), were generated from infusing dissolved drug (e.g., in ethanol) into the dissoln. medium and compared with that predicted from a comprehensive mechanistic model based on the classical nucleation theory taking into account both the particle growth and ripening processes. In the absence of any dissolved polymer to inhibit drug pptn., both our exptl. and predicted results show that the max. achievable supersatn. (i.e., kinetic soly.) of the amorphous solids increases, the time to reach max. decreases, and the rate of concn. decline in the de-supersatn. phase increases, with increasing rate of supersatn. generation (i.e., dissoln. rate). Our mechanistic model also predicts the existence of an optimal supersatn. rate which maximizes the area under the curve (AUC) of the kinetic soly. concn.-time profile, which agrees well with exptl. data. In the presence of a dissolved polymer from ASD dissoln., these obsd. trends also hold true except the de-supersatn. phase is more extended due to the crystn. inhibition effect. Since the obsd. kinetic soly. of nonequil. amorphous solids depends on the rate of supersatn. generation, our results also highlight the underlying difficulty in detg. a reproducible soly. advantage for amorphous solids.
- 44Zhu, L.; Wong, L.; Yu, L. Surface-enhanced crystallization of amorphous nifedipine. Mol. Pharmaceutics 2008, 5 (6), 921– 926, DOI: 10.1021/mp800063844https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtFChsbvE&md5=9f87dc51ab6f12c143b2760fa719346bSurface-Enhanced Crystallization of Amorphous NifedipineZhu, Lei; Wong, Letitia; Yu, LianMolecular Pharmaceutics (2008), 5 (6), 921-926CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Amorphous solids are generally more sol. and faster dissolving than their cryst. counterparts, a property useful for delivering poorly sol. drugs. Amorphous drugs must be stable against crystn., for crystn. negates their advantages. Recent studies found that crystal growth in amorphous indomethacin is orders of magnitude faster at the free surface than through the bulk and this surface-enhanced crystn. can be inhibited by an ultrathin coating. Herein, we report a second system that exhibits the same phenomena. Crystal growth at the free surface of amorphous nifedipine (NIF) was at least 1 order of magnitude faster than that through the bulk below the glass transition temp. Tg (42 °C). A thin coating of gold (10 nm) reduced the surface crystal growth rate to the bulk crystal growth rate. Surface-enhanced crystal growth was more pronounced near and below Tg than substantially above Tg, which suggests that this growth mode is more important for the glassy state. Our results support the view that a thin layer of mols. near the surface have higher mobility than the bulk mols. and can enable faster crystal growth. The higher mobility of surface mols. and the resulting fast crystal growth can be suppressed by an ultrathin coating.
- 45Hasebe, M.; Musumeci, D.; Yu, L. Fast surface crystallization of molecular glasses: creation of depletion zones by surface diffusion and crystallization flux. J. Phys. Chem. B 2015, 119 (7), 3304– 3311, DOI: 10.1021/jp512400c45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlyksro%253D&md5=893b871632f9b82d57a79d5140c69c47Fast Surface Crystallization of Molecular Glasses: Creation of Depletion Zones by Surface Diffusion and Crystallization FluxHasebe, Mariko; Musumeci, Daniele; Yu, LianJournal of Physical Chemistry B (2015), 119 (7), 3304-3311CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Mol. glasses can grow crystals much faster at the free surface than in the interior. A property of this process is the creation of depressed grooves or depletion zones around the crystals on the initially flat amorphous surface. With SEM and at. force microscopy, the authors studied this phenomenon in indomethacin (IMC), which crystallizes in two polymorphs (α and γ) of different morphologies. The obsd. depletion zones are well reproduced by the known coeffs. of surface diffusion and the velocities of crystal growth. At the slow-growing flanks of needle-like α-IMC crystals, depletion zones widen and deepen over time according to the expected kinetics for surface diffusion responding to a crystn. flux. Before fast-advancing growth fronts, depletion zones have less time to develop; their steady-state dimensions agree with the same model revised for a moving phase boundary. These results support the view that surface diffusion enables fast surface crystal growth on mol. glasses. The authors' finding helps understand crystal growth in thin films in which the formation of deep depletion zones can cause dewetting and alter growth kinetics.
- 46Zhu, L.; Jona, J.; Nagapudi, K.; Wu, T. Fast surface crystallization of amorphous griseofulvin below Tg. Pharm. Res. 2010, 27 (8), 1558– 1567, DOI: 10.1007/s11095-010-0140-846https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXltV2ksr8%253D&md5=e3740687002f83f51e2acfa5b5c7e759Fast Surface Crystallization of Amorphous Griseofulvin Below TgZhu, Lei; Jona, Janan; Nagapudi, Karthik; Wu, TianPharmaceutical Research (2010), 27 (8), 1558-1567CODEN: PHREEB; ISSN:0724-8741. (Springer)To study crystal growth rates of amorphous griseofulvin (GSF) below its glass transition temp. (Tg) and the effect of surface crystn. on the overall crystn. kinetics of amorphous GSF. Amorphous GSF was generated by melt quenching. Surface and bulk crystal growth rates were detd. using polarized light microscope. X-ray powder diffraction (XRPD) and Raman microscopy were used to identify the polymorph of the crystals. Crystn. kinetics of amorphous GSF powder stored at 40° (Tg-48°) and room temp. (Tg-66°) was monitored using XRPD. Results: Crystal growth at the surface of amorphous GSF is 10- to 100-fold faster than that in the bulk. The surface crystal growth can be suppressed by an ultrathin gold coating. Below Tg, the crystn. of amorphous GSF powder was biphasic with a rapid initial crystn. stage dominated by the surface crystn. and a slow or suspended late stage controlled by the bulk crystn. GSF exhibits the fastest surface crystn. kinetics among the known amorphous pharmaceutical solids. Well below Tg, surface crystn. dominated the overall crystn. kinetics of amorphous GSF powder. Thus, surface crystn. should be distinguished from bulk crystn. in studying, modeling and controlling the crystn. of amorphous solids.
- 47Elkhabaz, A.; Sarkar, S.; Dinh, J. K.; Simpson, G. J.; Taylor, L. S. Variation in supersaturation and phase behavior of ezetimibe amorphous solid dispersions upon dissolution in different biorelevant media. Mol. Pharmaceutics 2018, 15 (1), 193– 206, DOI: 10.1021/acs.molpharmaceut.7b0081447https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvVeisrvK&md5=fac144655adaafc768cbe00785514451Variation in Supersaturation and Phase Behavior of Ezetimibe Amorphous Solid Dispersions upon Dissolution in Different Biorelevant MediaElkhabaz, Ahmed; Sarkar, Sreya; Dinh, Janny K.; Simpson, Garth J.; Taylor, Lynne S.Molecular Pharmaceutics (2018), 15 (1), 193-206CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The delivery of poorly water-sol. drugs using amorphous solid dispersions (ASDs) has been widely acknowledged as a promising strategy for enhancing oral bioavailability. Upon dissoln., ASDs have accelerated dissoln. rates and yield supersatd. solns. leading to higher apparent solubilities. Understanding the complex phase behavior of ASDs during dissoln. is crucial for developing an effective formulation. Since the absorption of a lipophilic, high permeability drug is detd. primarily by the intraluminal dissoln. process and the final concn. achieved, there is a need for evaluation in biorelevant dissoln. media that simulate both fasting and fed gastrointestinal states. In this study, using ezetimibe as a model drug, three different ASDs were prepd. using poly(acrylic acid) (PAA), polyvinylpyrrolidone (PVP), and hydroxypropyl methylcellulose acetyl succinate (HPMC-AS). Dissoln. of ASDs was carried out in sodium phosphate buffer, fed-state simulated intestinal fluid (FeSSIF), and Ensure Plus to evaluate the impact of different dissoln. media on release profile, supersatn., and phase behavior. The supersatn. level and crystn. kinetics varied among the dispersions and were found to be highly dependent on the medium employed. The presence of solubilizing additives in biorelevant media greatly affected the generation and stabilization of supersatd. solns. Second harmonic generation microscopy was found to enable the detection of crystals in all media including the highly turbid Ensure Plus system. In conclusion, it is important to evaluate the impact of complex biorelevant media on the dissoln. performance of ASDs to better design supersaturating formulations for oral delivery.
Supporting Information
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.molpharmaceut.8b00840.
ATR-FTIR spectra of tablet surfaces after dissolution and additional CARS and SFG overlay images of tablets stored for 1 and 7 days at 30 °C/75% RH (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.