Skip to main content
Log in

Densifying Co-Precipitated Amorphous Dispersions to Achieve Improved Bulk Powder Properties

  • Original Research Article
  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

Purpose

Precipitation of amorphous solid dispersions has gained traction in the pharmaceutical industry given its application to pharmaceuticals with varying physicochemical properties. Although preparing co-precipitated amorphous dispersions (cPAD) in high-shear rotor–stator devices allows for controlled shear conditions during precipitation, such aggressive mixing environments can result in materials with low bulk density and poor flowability. This work investigated annealing cPAD after precipitation by washing with heated anti-solvent to improve bulk powder properties required for downstream drug product processing.

Methods

Co-precipitation dispersions were prepared by precipitation into pH-modified aqueous anti-solvent. Amorphous dispersions were washed with heated anti-solvent and assessed for bulk density, flowability, and dissolution behavior relative to both cPAD produced without a heated wash and spray dried intermediate.

Results

Washing cPAD with a heated anti-solvent resulted in an improvement in flowability and increased bulk density. The mechanism of densification was ascribed to annealing over the wetted Tg of the material, which lead to collapse of the porous co-precipitate structure into densified granules without causing crystallization. In contrast, an alternative approach to increase bulk density by precipitating the ASD using low shear conditions showed evidence of crystallinity. The dissolution rate of the densified cPAD granules was lower than that of the low-bulk density dispersions, although both samples reached concentrations equivalent to that of the spray dried intermediate after 90 min dissolution.

Conclusions

Hot wash densification was a tenable route to produce co-precipitated amorphous dispersions with improved properties for downstream processing compared to non-densified powders.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Baghel S, Cathcart H, O’Reilly NJ. Polymeric amorphous solid dispersions: a review of amorphization, crystallization, stabilization, solid-state characterization, and aqueous solubilization of biopharmaceutical classification system class II drugs. J Pharm Sci. 2016;105(9):2527–44.

    Article  CAS  Google Scholar 

  2. Shah N, Sandhu H, Choi DS, Chokshi H, Malick AW. Amorphous solid dispersions Theory and Practice. Berlin, Germany: Springer; 2014.

    Book  Google Scholar 

  3. Yu L. Amorphous pharmaceutical solids: preparation, characterization and stabilization. Adv Drug Deliv Rev. 2001;48(1):27–42.

    Article  CAS  Google Scholar 

  4. Jermain SV, Brough C, Williams RO III. Amorphous solid dispersions and nanocrystal technologies for poorly water-soluble drug delivery–an update. Int J Pharm. 2018;535(1–2):379–92.

    Article  CAS  Google Scholar 

  5. Moseson DE, Taylor LS. The application of temperature-composition phase diagrams for hot melt extrusion processing of amorphous solid dispersions to prevent residual crystallinity. Int J Pharm. 2018;553(1–2):454–66.

    Article  CAS  Google Scholar 

  6. LaFountaine JS, McGinity JW, Williams RO. Challenges and strategies in thermal processing of amorphous solid dispersions: a review. AAPS PharmSciTech. 2016;17(1):43–55.

    Article  CAS  Google Scholar 

  7. Ziaee A, Albadarin AB, Padrela L, Faucher A, O’Reilly E, Walker G. Spray drying ternary amorphous solid dispersions of ibuprofen–An investigation into critical formulation and processing parameters. Eur J Pharm Biopharm. 2017;120:43–51.

    Article  CAS  Google Scholar 

  8. Williams HD, Trevaskis NL, Charman SA, Shanker RM, Charman WN, Pouton CW, et al. Strategies to address low drug solubility in discovery and development. Pharmacol Rev. 2013;65(1):315–499.

    Article  Google Scholar 

  9. Lee Y-C, McNevin M, Ikeda C, Chouzouri G, Moser J, Harris D, et al. Combination of colloidal silicon dioxide with spray-dried solid dispersion to facilitate discharge from an agitated dryer. AAPS PharmSciTech. 2019;20(5):1–7.

    Article  Google Scholar 

  10. Poozesh S, Setiawan N, Arce F, Sundararajan P, Della Rocca J, Rumondor A, et al. Understanding the process-product-performance interplay of spray dried drug-polymer systems through complete structural and chemical characterization of single spray dried particles. Powder Technol. 2017;320:685–95.

    Article  CAS  Google Scholar 

  11. Honick M, Das S, Hoag SW, Muller FX, Alayoubi A, Feng X, et al. The effects of spray drying, HPMCAS grade, and compression speed on the compaction properties of itraconazole-HPMCAS spray dried dispersions. Eur J Pharm Sci. 2020;155: 105556.

    Article  CAS  Google Scholar 

  12. Patel S, Kou X, Hou HH, Huang YB, Strong JC, Zhang GG, et al. Mechanical properties and tableting behavior of amorphous solid dispersions. J Pharm Sci. 2017;106(1):217–23.

    Article  CAS  Google Scholar 

  13. Démuth B, Nagy ZK, Balogh A, Vigh T, Marosi G, Verreck G, et al. Downstream processing of polymer-based amorphous solid dispersions to generate tablet formulations. Int J Pharm. 2015;486(1–2):268–86.

    Article  Google Scholar 

  14. Ekdahl A, Mudie D, Malewski D, Amidon G, Goodwin A. Effect of spray-dried particle morphology on mechanical and flow properties of felodipine in PVP VA amorphous solid dispersions. J Pharm Sci. 2019;108(11):3657–66.

    Article  CAS  Google Scholar 

  15. Brown C, DiNunzio J, Eglesia M, Forster S, Lamm M, Lowinger M, et al. Hot-melt extrusion for solid dispersions: composition and design considerations. Amorphous Solid Dispersions: Springer; 2014. p. 197–230.

    Google Scholar 

  16. Breitenbach J. Melt extrusion: from process to drug delivery technology. Eur J Pharm Biopharm. 2002;54(2):107–17.

    Article  CAS  Google Scholar 

  17. Schenck LR, Lamberto DJ, Kukura IJL, Guzman FJ, Cote A, Koynov A. Process for preparing pharmaceutical compositions. United States patent application US 16/061,513. 2020.

  18. Hou HH, Rajesh A, Pandya KM, Lubach JW, Muliadi A, Yost E, et al. Impact of method of preparation of amorphous solid dispersions on mechanical properties: Comparison of coprecipitation and spray drying. J Pharm Sci. 2019;108(2):870–9.

    Article  CAS  Google Scholar 

  19. Sturm DR, Moser JD, Sundararajan P, Danner RP. Spray drying of hypromellose acetate succinate. Ind Eng Chem Res. 2019;58(27):12291–300.

    Article  CAS  Google Scholar 

  20. Al-Khattawi A, Bayly A, Phillips A, Wilson D. The design and scale-up of spray dried particle delivery systems. Expert Opin Drug Deliv. 2018;15(1):47–63.

    Article  CAS  Google Scholar 

  21. Poozesh S, Mahdi JS. Are traditional small-scale screening methods reliable to predict pharmaceutical spray drying? Pharm Dev Technol. 2019;24(7):915–25.

    Article  CAS  Google Scholar 

  22. Dong Z, Chatterji A, Sandhu H, Choi DS, Chokshi H, Shah N. Evaluation of solid state properties of solid dispersions prepared by hot-melt extrusion and solvent co-precipitation. Int J Pharm. 2008;355(1–2):141–9.

    Article  CAS  Google Scholar 

  23. Simonelli A, Mehta S, Higuchi W. Dissolution rates of high energy sulfathiazole-povidone coprecipitates II: characterization of form of drug controlling its dissolution rate via solubility studies. J Pharm Sci. 1976;65(3):355–61.

    Article  CAS  Google Scholar 

  24. Sertsou G, Butler J, Hempenstall J, Rades T. Solvent change co-precipitation with hydroxypropyl methylcellulose phthalate to improve dissolution characteristics of a poorly water-soluble drug. J Pharm Pharmacol. 2002;54(8):1041–7.

    Article  CAS  Google Scholar 

  25. Schenck L, Boyce C, Frank D, Koranne S, Ferguson HM, Strotman N. Hierarchical Particle Approach for Co-Precipitated Amorphous Solid Dispersions for Use in Preclinical In Vivo Studies. Pharmaceutics. 2021;13(7):1034.

    Article  CAS  Google Scholar 

  26. Jia W, Yawman PD, Pandya KM, Sluga K, Ng T, Kou D, et al. Assessing the interrelationship of microstructure, properties, drug release performance, and preparation process for amorphous solid dispersions via noninvasive imaging analytics and material characterization. Pharma Res. 2022:1–18.

  27. Song S, Wang C, Wang S, Siegel RA, Sun CC. Efficient development of sorafenib tablets with improved oral bioavailability enabled by coprecipitated amorphous solid dispersion. Int J Pharma. 2021;610:121216.

  28. Shah N, Iyer RM, Mair H-J, Choi D, Tian H, Diodone R, et al. Improved human bioavailability of vemurafenib, a practically insoluble drug, using an amorphous polymer-stabilized solid dispersion prepared by a solvent-controlled coprecipitation process. J Pharm Sci. 2013;102(3):967–81.

    Article  CAS  Google Scholar 

  29. Frank D, Schenck L, Koynov A, Su Y, Li Y, Variankaval N. Optimizing Solvent Selection and Processing Conditions to Generate High Bulk-Density, Co-Precipitated Amorphous Dispersions of Posaconazole. Pharmaceutics. 2021;13(12):2017.

    Article  CAS  Google Scholar 

  30. Frank D, Nie H, Chandra A, Coelho A, Dalton C, Dvorak H, et al. High bulk-density amorphous dispersions to enable direct compression of reduced image size amorphous dosage units. J Pharma Sci. 2022. in press.

  31. Schenck L, Koynov A, Cote A. Particle engineering at the drug substance, drug product interface: a comprehensive platform approach to enabling continuous drug substance to drug product processing with differentiated material properties. Drug Dev Ind Pharm. 2019;45(4):521–31.

    Article  CAS  Google Scholar 

  32. Strotman NA, Schenck L. Coprecipitated amorphous dispersions as drug substance: opportunities and challenges. Org Process Res Dev. 2022;26(1):10–3.

  33. Matteucci ME, Hotze MA, Johnston KP, Williams RO. Drug nanoparticles by antisolvent precipitation: mixing energy versus surfactant stabilization. Langmuir. 2006;22(21):8951–9.

    Article  CAS  Google Scholar 

  34. Harter A, Schenck L, Lee I, Cote A. High-shear rotor–stator wet milling for drug substances: expanding capability with improved scalability. Org Process Res Dev. 2013;17(10):1335–44.

    Article  CAS  Google Scholar 

  35. Schenck L, Mann AKP, Liu Z, Milewski M, Zhang S, Ren J, et al. Building a better particle: Leveraging physicochemical understanding of amorphous solid dispersions and a hierarchical particle approach for improved delivery at high drug loadings. Int J Pharm. 2019;559:147–55.

    Article  CAS  Google Scholar 

  36. Gamble JF, Chiu W-S, Tobyn M. Investigation into the impact of sub-populations of agglomerates on the particle size distribution and flow properties of conventional microcrystalline cellulose grades. Pharm Dev Technol. 2011;16(5):542–8.

    Article  CAS  Google Scholar 

  37. Horio T, Yasuda M, Matsusaka S. Effect of particle shape on powder flowability of microcrystalline cellulose as determined using the vibration shear tube method. Int J Pharm. 2014;473(1–2):572–8.

    Article  CAS  Google Scholar 

  38. Barjat H, Checkley S, Chitu T, Dawson N, Farshchi A, Ferreira A, et al. Demonstration of the feasibility of predicting the flow of pharmaceutically relevant powders from particle and bulk physical properties. J Pharm Innov. 2021;16(1):181–96.

    Article  Google Scholar 

  39. Bhujbal SV, Pathak V, Zemlyanov DY, Taylor LS, Zhou QT. Physical stability and dissolution of lumefantrine amorphous solid dispersions produced by spray anti-solvent precipitation. J Pharm Sci. 2021;110(6):2423–31.

    Article  CAS  Google Scholar 

  40. Ikeda C, Zhou G, Lee Y-C, Chouzouri G, Howell L, Marshall B, et al. Application of online NIR spectroscopy to enhance process understanding and enable in-process control testing of secondary drying process for a spray-dried solid dispersion intermediate. J Pharma Sci. 2022;111(9):2540–51.

  41. Lowinger M, Baumann J, Vodak DT, Moser J. Practical considerations for spray dried formulation and process development. Discovering and developing molecules with optimal drug-like properties: Springer; 2015. p. 383–435.

    Google Scholar 

  42. Cote A, Sirota E. CRYSTALLIZATION: the pursuit of a robust approach for growing crystals directly to target size. American Pharmaceutical Review. 2010;13(7):46.

    Google Scholar 

  43. Meng W, Sirota E, Feng H, McMullen JP, Codan L, Cote AS. Effective Control of Crystal Size via an Integrated Crystallization, Wet Milling, and Annealing Recirculation System. Org Process Res Dev. 2020;24(11):2639–50.

    Article  CAS  Google Scholar 

  44. Hancock BC, Zografi G. Characteristics and significance of the amorphous state in pharmaceutical systems. J Pharm Sci. 1997;86(1):1–12.

    Article  CAS  Google Scholar 

  45. Debenedetti PG, Stillinger FH. Supercooled liquids and the glass transition. Nature. 2001;410(6825):259–67.

    Article  CAS  Google Scholar 

  46. Krishna Kumar N, Suryanarayanan R. Crystallization propensity of amorphous pharmaceuticals: kinetics and thermodynamics. Mol Pharma. 2022;19(2):472–83.

  47. Indulkar AS, Lou X, Zhang GG, Taylor LS. Insights into the dissolution mechanism of ritonavir–copovidone amorphous solid dispersions: importance of congruent release for enhanced performance. Mol Pharm. 2019;16(3):1327–39.

    Article  CAS  Google Scholar 

  48. Newman A, Zografi G. What are the important factors that influence API crystallization in miscible amorphous API–excipient mixtures during long-term storage in the glassy state? Mol Pharma. 2021;19(2):378–91.

  49. Friesen DT, Shanker R, Crew M, Smithey DT, Curatolo W, Nightingale J. Hydroxypropyl methylcellulose acetate succinate-based spray-dried dispersions: an overview. Mol Pharm. 2008;5(6):1003–19.

    Article  CAS  Google Scholar 

  50. Wuelfing WP, El Marrouni A, Lipert MP, Daublain P, Kesisoglou F, Converso A, et al. Dose number as a tool to guide lead optimization for orally bioavailable compounds in drug discovery. J Med Chem. 2022;65(3):1685–94.

Download references

Acknowledgements

The authors acknowledge Steve Crowley, Jim DiNunzio, Edi Meco, and Graciela Terife for work producing the Compound X SDI, Sebastian Escotet, Prapti Kafle, Grace Okoh, Andrew Parker, and Laura Wareham for engaging discussions in the preparation of this work, and Erin Guidry, Joe Kukura, Matthew Lamm, Ian Mangion, Becky Ruck, and Neil Strotman for support.

Funding

This work was funded by Merck & Co., Inc., Rahway, NJ, USA.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Derek S. Frank.

Ethics declarations

Conflicts of Interest

The authors declare no conflicts of interest. The authors are employees of Merck & Co. Inc, Rahway, NJ, USA. Merck & Co. Inc. had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 14500 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Frank, D.S., Punia, A., Fahy, M. et al. Densifying Co-Precipitated Amorphous Dispersions to Achieve Improved Bulk Powder Properties. Pharm Res 39, 3197–3208 (2022). https://doi.org/10.1007/s11095-022-03416-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11095-022-03416-6

Keywords

Navigation