Skip to main content

Advertisement

Log in

The Influence of Cellulosic Polymer’s Variables on Dissolution/Solubility of Amorphous Felodipine and Crystallization Inhibition from a Supersaturated State

  • Research Article
  • Published:
AAPS PharmSciTech Aims and scope Submit manuscript

Abstract

The collective impact of cellulosic polymers on the dissolution, solubility, and crystallization inhibition of amorphous active pharmaceutical ingredients (APIs) is still far from being adequately understood. The goal of this research was to explore the influence of cellulosic polymers and incubation conditions on enhancement of solubility and dissolution of amorphous felodipine, while inhibiting crystallization of the drug from a supersaturated state. Variables, including cellulosic polymer type, amount, ionic strength, and viscosity, were evaluated for effects on API dissolution/solubility and crystallization processes. Water-soluble cellulosic polymers, including HPMC E15, HPMC E5, HPMC K100-LV, L-HPC, and MC, were studied. All cellulosic polymers could extend API dissolution and solubility to various extents by delaying crystallization and prolonging supersaturation duration, with their effectiveness ranked from greatest to least as HPMC E15 > HPMC E5 > HPMC K100-LV > L-HPC > MC. Decreased polymer amount, lower ionic strength, or higher polymer viscosity tended to decrease dissolution/solubility and promote crystal growth to accelerate crystallization. HPMC E15 achieved greatest extended API dissolution and maintenance of supersaturation from a supersaturated state; this polymer thus had the greatest potential for maintaining sustainable API absorption within biologically relevant time frames.

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
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11

Similar content being viewed by others

References

  1. Amidon GL, Lennernäs H, Shah VP, Crison JR. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res. 1995;12(3):413–20.

    Article  CAS  PubMed  Google Scholar 

  2. Khadra I, Zhou Z, Dunn C, Wilson CG, Halbert G. Statistical investigation of simulated intestinal fluid composition on the equilibrium solubility of biopharmaceutics classification system class II drugs. Eur J Pharm Sci. 2015;67:65–75.

    Article  CAS  PubMed  Google Scholar 

  3. Xie F, Ji S, Cheng Z. In vitro dissolution similarity factor (f2) and in vivo bioequivalence criteria, how and when do they match? Using a BCS class II drug as a simulation example. Eur J Pharm Sci. 2015;66:163–72.

    Article  CAS  PubMed  Google Scholar 

  4. Han X, Ghoroi C, To D, Chen Y, Davé R. Simultaneous micronization and surface modification for improvement of flow and dissolution of drug particles. Int J Pharm. 2011;415(1–2):185–95.

    Article  CAS  PubMed  Google Scholar 

  5. He Y, Orton E, Yang D. The selection of a pharmaceutical salt-the effect of the acidity of the counterion on its solubility and potential biopharmaceutical performance. J Pharm Sci. 2018;107(1):419–25.

    Article  CAS  PubMed  Google Scholar 

  6. Zimmermann SC, Tichý T, Vávra J, Dash RP, Slusher CE, Gadiano AJ, et al. N-substituted prodrugs of mebendazole provide improved aqueous solubility and oral bioavailability in mice and dogs. J Med Chem. 2018;61(9):3918–29.

    Article  CAS  PubMed  Google Scholar 

  7. Jansook P, Ogawa N, Loftsson T. Cyclodextrins: structure, physicochemical properties and pharmaceutical applications. Int J Pharm. 2018;535(1–2):272–84.

    Article  CAS  PubMed  Google Scholar 

  8. Parikh T, Serajuddin ATM. Development of fast-dissolving amorphous solid dispersion of itraconazole by melt extrusion of its mixture with weak organic carboxylic acid and polymer. Pharm Res. 2018;35(7):127.

    Article  PubMed  CAS  Google Scholar 

  9. Healy AM, Worku ZA, Kumar D, Madi AM. Pharmaceutical solvates, hydrates and amorphous forms: a special emphasis on cocrystals. Adv Drug Deliv Rev. 2017;117:25–46.

    Article  CAS  PubMed  Google Scholar 

  10. Edueng K, Mahlin D, Bergström CAS. The need for restructuring the disordered science of amorphous drug formulations. Pharm Res. 2017;34(9):1754–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kissi EO, Grohganz H, Löbmann K, Ruggiero MT, Zeitler JA, Rades T. Glass-transition temperature of the β-relaxation as the major predictive parameter for recrystallization of neat amorphous drugs. J Phys Chem B. 2018;122(10):2803–8.

    Article  CAS  PubMed  Google Scholar 

  12. Doreth M, Löbmann K, Priemel P, Grohganz H, Taylor R, Holm R, et al. Influence of PVP molecular weight on the microwave assisted in situ amorphization of indomethacin. Eur J Pharm Biopharm. 2018;122:62–9.

    Article  CAS  PubMed  Google Scholar 

  13. Lehmkemper K, Kyeremateng SO, Heinzerling O, Degenhardt M, Sadowski G. Long-term physical stability of PVP- and PVPVA-amorphous solid dispersions. Mol Pharm. 2017;14(1):157–71.

    Article  CAS  PubMed  Google Scholar 

  14. Li N, Taylor LS. Tailoring supersaturation from amorphous solid dispersions. J Control Release. 2018;279:114–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Van den Mooter G. The use of amorphous solid dispersions: a formulation strategy to overcome poor solubility and dissolution rate. Drug Discov Today Technol. 2012;9(2):e71–e174.

    Article  CAS  Google Scholar 

  16. Lehmkemper K, Kyeremateng SO, Bartels M, Degenhardt M, Sadowski G. Physical stability of API/polymer-blend amorphous solid dispersions. Eur J Pharm Biopharm. 2018;124:147–57.

    Article  CAS  PubMed  Google Scholar 

  17. Dos Santos KM, Barbosa RM, Vargas FGA, de Azevedo EP, Lins ACDS, Camara CA, et al. Development of solid dispersions of β-lapachone in PEG and PVP by solvent evaporation method. Drug Dev Ind Pharm. 2018;44(5):750–6.

    Article  CAS  PubMed  Google Scholar 

  18. Shi NQ, Lei YS, Song LM, Zhang XB, Wang XL. Impact of amorphous and semicrystalline polymers on the dissolution and crystallization inhibition of pioglitazone solid dispersions. Powder Technol. 2013;247:211–21.

    Article  CAS  Google Scholar 

  19. Miyazaki T, Yoshioka S, Aso Y, Kojima S. Ability of polyvinylpyrrolidone and polyacrylic acid to inhibit the crystallization of amorphous acetaminophen. J Pharm Sci. 2004;93(11):2710–7.

    Article  CAS  PubMed  Google Scholar 

  20. Van den Mooter G, Wuyts M, Blaton N, Busson R, Grobet P, Augustijns P, et al. Physical stabilization of amorphous ketoconazole in solid dispersions with polyvinylpyrrolidone K25. Eur J Pharm Sci. 2001;12(3):261–9.

    Article  PubMed  Google Scholar 

  21. Aso Y, Yoshioka S, Zhang J, Zografi G. Effect of water on the molecular mobility of sucrose and poly(vinylpyrrolidone) in a colyophilized formulation as measured by (13)C-NMR relaxation time. Chem Pharm Bull. 2002;50(6):822–6.

    Article  CAS  PubMed  Google Scholar 

  22. Alonzo DE, Gao Y, Zhou D, Mo H, Zhang GGZ, Taylor LS. Dissolution and precipitation behavior of amorphous solid dispersions. J Pharm Sci. 2011;100(8):3316–31.

    Article  CAS  PubMed  Google Scholar 

  23. Sugihara H, Taylor LS. Evaluation of pazopanib phase behavior following pH-induced supersaturation. Mol Pharm. 2018;15(4):1690–9.

    Article  CAS  PubMed  Google Scholar 

  24. Xie T, Gao W, Taylor LS. Impact of Eudragit EPO and hydroxypropyl methylcellulose on drug release rate, supersaturation, precipitation outcome and redissolution rate of indomethacin amorphous solid dispersions. Int J Pharm. 2017;531(1):313–23.

    Article  CAS  PubMed  Google Scholar 

  25. Sarode AL, Wang P, Obara S, Worthen DR. Supersaturation, nucleation, and crystal growth during single- and biphasic dissolution of amorphous solid dispersions: polymer effects and implications for oral bioavailability enhancement of poorly water soluble drugs. Eur J Pharm Biopharm. 2014;86(3):351–60.

    Article  CAS  PubMed  Google Scholar 

  26. Konno H, Handa T, Alonzo DE, Taylor LS. Effect of polymer type on the dissolution profile of amorphous solid dispersions containing felodipine. Eur J Pharm Biopharm. 2008;70(2):493–9.

    Article  CAS  PubMed  Google Scholar 

  27. Tran TTD, Tran PHL. Perspectives on strategies using swellable polymers in solid dispersions for controlled drug release. Curr Pharm Des. 2017;23(11):1639–48.

    Article  CAS  PubMed  Google Scholar 

  28. Van Duong T, Van den Mooter G. The role of the carrier in the formulation of pharmaceutical solid dispersions. Part II: amorphous carriers. Expert Opin Drug Deliv. 2016;13(12):1681–94.

    Article  PubMed  CAS  Google Scholar 

  29. van der Lee R, Pfaffendorf M, Koopmans RP, van Lieshout JJ, van Montfrans GA, van Zwieten PA. Comparison of the time courses and potencies of the vasodilator effects of nifedipine and felodipine in the human forearm. Blood Press. 2001;10(4):217–22.

    Article  PubMed  Google Scholar 

  30. Leenen FH, Coletta E. Pharmacokinetic and antihypertensive profile of amlodipine and felodipine-ER in younger versus older patients with hypertension. J Cardiovasc Pharmacol. 2010;56(6):669–75.

    Article  CAS  PubMed  Google Scholar 

  31. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2001;46(1–3):3–26.

    Article  CAS  PubMed  Google Scholar 

  32. Hancock BC, Parks M. What is the true solubility advantage for amorphous pharmaceuticals? Pharm Res. 2000;17(4):397–404.

    Article  CAS  PubMed  Google Scholar 

  33. Bikiaris DN. Solid dispersions, part II: new strategies in manufacturing methods for dissolution rate enhancement of poorly water-soluble drugs. Expert Opin Drug Deliv. 2011;8(12):1663–80.

    Article  CAS  PubMed  Google Scholar 

  34. Shi NQ, Lai HW, Zhang Y, Feng B, Xiao X, Zhang HM, et al. On the inherent properties of Soluplus and its application in ibuprofen solid dispersions generated by microwave-quench cooling technology. Pharm Dev Technol. 2018;23(6):573–86.

    Article  CAS  PubMed  Google Scholar 

  35. Tran PH, Tran TT, Lee KH, Kim DJ, Lee BJ. Dissolution-modulating mechanism of pH modifiers in solid dispersion containing weakly acidic or basic drugs with poor water solubility. Expert Opin Drug Deliv. 2010;7(5):647–61.

    Article  CAS  PubMed  Google Scholar 

  36. Fong SY, Bauer-Brandl A, Brandl M. Oral bioavailability enhancement through supersaturation: an update and meta-analysis. Expert Opin Drug Deliv. 2017;14(3):403–26.

    Article  CAS  PubMed  Google Scholar 

  37. Laitinen R, Löbmann K, Grohganz H, Priemel P, Strachan CJ, Rades T. Supersaturating drug delivery systems: the potential of co-amorphous drug formulations. Int J Pharm. 2017;532(1):1–12.

    Article  CAS  PubMed  Google Scholar 

  38. Brouwers J, Brewster ME, Augustijns P. Supersaturating drug delivery systems: the answer to solubility-limited oral bioavailability? J Pharm Sci. 2009;98(8):2549–72.

    Article  CAS  PubMed  Google Scholar 

  39. Brewster ME, Vandecruys R, Verreck G, Peeters J. Supersaturating drug delivery systems: effect of hydrophilic cyclodextrins and other excipients on the formation and stabilization of supersaturated drug solutions. Pharmazie. 2008;63(3):217–20.

    CAS  PubMed  Google Scholar 

  40. Guzmán HR, Tawa M, Zhang Z, Ratanabanangkoon P, Shaw P, Gardner CR, et al. Combined use of crystalline salt forms and precipitation inhibitors to improve oral absorption of celecoxib from solid oral formulations. J Pharm Sci. 2007;96(10):2686–702.

    Article  PubMed  CAS  Google Scholar 

  41. Kawakami K. Modification of physicochemical characteristics of active pharmaceutical ingredients and application of supersaturatable dosage forms for improving bioavailability of poorly absorbed drugs. Adv Drug Deliv Rev. 2012;64(6):480–95.

    Article  CAS  PubMed  Google Scholar 

  42. Shi NQ, Zhang Y, Li Y, Lai HW, Xiao X, Feng B, et al. Self-micellizing solid dispersions enhance the properties and therapeutic potential of fenofibrate: advantages, profiles and mechanisms. Int J Pharm. 2017;528(1–2):563–77.

    Article  CAS  PubMed  Google Scholar 

  43. Xu S, Dai WG. Drug precipitation inhibitors in supersaturable formulations. Int J Pharm. 2013;453(1):36–43.

    Article  CAS  PubMed  Google Scholar 

  44. Liu C, Chen Z, Chen Y, Lu J, Li Y, Wang S, et al. Improving oral bioavailability of sorafenib by optimizing the "spring" and "parachute" based on molecular interaction mchanisms. Mol Pharm. 2016;13(2):599–608.

    Article  CAS  PubMed  Google Scholar 

  45. Sun DD, Lee PI. Evolution of supersaturation of amorphous pharmaceuticals: nonlinear rate of supersaturation generation regulated by matrix diffusion. Mol Pharm. 2015;12(4):1203–15.

    Article  CAS  PubMed  Google Scholar 

  46. Turnbull D, Fischer JC. Rate of nucleation in condensed systems. J Chem. Physics. 1949;17:71–3.

    CAS  Google Scholar 

  47. Schver GCRM, Lee PI. Combined effects of supersaturation rates and doses on the kinetic-solubility profiles of amorphous solid dispersions based on water-insoluble poly(2-hydroxyethyl methacrylate) hydrogels. Mol Pharm. 2018;15(5):2017–26.

    Article  CAS  PubMed  Google Scholar 

  48. Taylor LS, Zhang GGZ. Physical chemistry of supersaturated solutions and implications for oral absorption. Adv Drug Deliv Rev. 2016;101:122–42.

    Article  CAS  PubMed  Google Scholar 

  49. Ueda K, Higashi K, Yamamoto K, Moribe K. Equilibrium state at supersaturated drug concentration achieved by hydroxypropyl methylcellulose acetate succinate: molecular characterization using (1)H NMR technique. Mol Pharm. 2015;12(4):1096–104.

    Article  CAS  PubMed  Google Scholar 

  50. Ueda K, Higashi K, Kataoka M, Yamashita S, Yamamoto K, Moribe K. Inhibition mechanism of hydroxypropyl methylcellulose acetate succinate on drug crystallization in gastrointestinal fluid and drug permeability from a supersaturated solution. Eur J Pharm Sci. 2014;62:293–300.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by Scientific Research Foundation of the Education Department of Jilin Province of China (No. 2015-401), Science and Technology Development Program of Jilin Province of China (Nos. 20160520046JH and 20140203012YY), the National Natural Science Foundation of China (No. 81602654), Science and Technology Development Program of Jilin City in Jilin Province of China (No. 201464053), the State Key Laboratory of Medicinal Chemical Biology of Nankai University (No. 201503001), and China Postdoctoral Science Foundation (No. 2015M571373).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Nian-Qiu Shi or Bo Feng.

Additional information

Publisher’s Note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shi, NQ., Jin, Y., Zhang, Y. et al. The Influence of Cellulosic Polymer’s Variables on Dissolution/Solubility of Amorphous Felodipine and Crystallization Inhibition from a Supersaturated State. AAPS PharmSciTech 20, 12 (2019). https://doi.org/10.1208/s12249-018-1266-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1208/s12249-018-1266-y

KEY WORDS

Navigation