Skip to main content
Log in

Investigating a Novel Hot Melt Extrusion-Based Drying Technique to Solidify an Amorphous Nanosuspension Using Design of Experiment Methodology

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

Abstract

The hot melt extrusion (HME) technology was explored and optimized to solidify an amorphous nanosuspension using Quality by Design (QbD) methodology. A design of experiments (DoE) approach was used to perform a set of 15 experiments, varying independent variables (feed rate, input temperature, and screw speed) within a design space. Redispersibility index (RDI), moisture content, and process yield constituted the critical quality attributes (CQAs) of the experimental design. Regression analysis and ANOVA were employed to identify and estimate significant main effects and two-way interactions, and model the process of HME drying for predictive purposes. The optimized HME-dried end product was characterized for physicochemical properties using differential scanning calorimetry (DSC), X-ray powder diffractions (XRPD), polarized light microscopy (PLM), Fourier transform infrared spectroscopy (FTIR), and in vitro dissolution studies. The statistical analysis reveals feed rate and input temperature as significant independent variables, critically influencing RDI and moisture content of solidified end product. The model developed for process yield was insignificant at a p-value of 0.05. The API retained its amorphous nature after the extrusion process which was confirmed using DSC and XRPD techniques. PLM was unsuitable to differentiate and determine crystallinity of drug moiety in the presence of a semi-crystalline bulking agent, microcrystalline cellulose (MCC). In vitro dissolution study depicted solubility and dissolution enhancement for HME-dried amorphous nanosuspension in both the dissolution media which can be attributed to amorphous nature of nanosized drug particles. A well-designed study implemented by DoE aided in developing a robust and novel HME technique to dry aqueous nanosuspension.

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11
Fig. 12
Fig. 13
Fig. 14

Similar content being viewed by others

References

  1. Pu X, Sun J, Li M, He Z. Formulation of nanosuspensions as a new approach for the delivery of poorly soluble drugs. Curr Nanosci. 2009;5(4):417–27.

    Article  CAS  Google Scholar 

  2. Babu NJ, Nangia A. Solubility advantage of amorphous drugs and pharmaceutical cocrystals. Cryst Growth Des. 2011;11(7):2662–79.

    Article  CAS  Google Scholar 

  3. Desai C, Prabhakar B. Nano-amorphous composites of cilostazol–HP-β-CD inclusion complexes: physicochemical characterization, structure elucidation, thermodynamic studies and in vitro evaluation. J Incl Phenom Macrocycl Chem. 2015;81(1–2):175–91.

    Article  CAS  Google Scholar 

  4. Memişoğlu E, Bochot A, Özalp M, Şen M, Duchêne D, Hincal AA. Direct formation of nanospheres from amphiphilic β-cyclodextrin inclusion complexes. Pharm Res. 2003;20(1):117–25.

    Article  PubMed  Google Scholar 

  5. Patel AA. Liquid salt as green solvent: a novel eco-friendly technique to enhance solubility and stability of poorly soluble drugs: Long Island University, The Brooklyn Center. 2015.

  6. Vogt M, Kunath K, Dressman JB. Dissolution enhancement of fenofibrate by micronization, cogrinding and spray-drying: comparison with commercial preparations. Eur J Pharm Biopharm. 2008;68(2):283–8.

    Article  CAS  PubMed  Google Scholar 

  7. Shah DA. To understand the thermodynamic and kinetic properties of nanocrystals using poorly soluble drugs: Long Island University, The Brooklyn Center. 2015.

  8. Thakuria R, Delori A, Jones W, Lipert MP, Roy L, Rodríguez-Hornedo N. Pharmaceutical cocrystals and poorly soluble drugs. Int J Pharm. 2013;453(1):101–25.

    Article  CAS  PubMed  Google Scholar 

  9. Dugar RP, Gajera BY, Dave RH. Fusion method for solubility and dissolution rate enhancement of ibuprofen using block copolymer poloxamer 407. AAPS PharmSciTech. 2016;17(6):1428–40.

    Article  CAS  PubMed  Google Scholar 

  10. Tank D, Karan K, Gajera BY, Dave RH. Investigate the effect of solvents on wet granulation of microcrystalline cellulose using hydroxypropyl methylcellulose as a binder and evaluation of rheological and thermal characteristics of granules. Saudi Pharm J. 2018;26(4):593–602.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Brough C, Williams Iii R. Amorphous solid dispersions and nano-crystal technologies for poorly water-soluble drug delivery. Int J Pharm. 2013;453(1):157–66.

    Article  CAS  PubMed  Google Scholar 

  12. Murdande SB, Pikal MJ, Shanker RM, Bogner RH. Solubility advantage of amorphous pharmaceuticals: I. A thermodynamic analysis. J Pharm Sci. 2010;99(3):1254–64.

    Article  CAS  PubMed  Google Scholar 

  13. Bates S, Zografi G, Engers D, Morris K, Crowley K, Newman A. Analysis of amorphous and nanocrystalline solids from their X-ray diffraction patterns. Pharm Res. 2006;23(10):2333–49.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. Laitinen R, Löbmann K, Strachan CJ, Grohganz H, Rades T. Emerging trends in the stabilization of amorphous drugs. Int J Pharm. 2013;453(1):65–79.

    Article  CAS  PubMed  Google Scholar 

  16. 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;532:1–12.

    Article  CAS  PubMed  Google Scholar 

  17. 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  PubMed  Google Scholar 

  18. Chaudhari SP, Dave RH. Evaluating the effects of different molecular weights of polymers in stabilizing supersaturated drug solutions and formulations using various methodologies of the model drug: fenofibrate. J Pharm Sci Pharmacol. 2015;2(3):259–76.

    Article  Google Scholar 

  19. Ruddy SB, Eickhoff WM, Liversidge G. Site-specific adhesion within the GI tract using nanoparticles stabilized by high molecular weight, linear poly (ethylene oxide) polymers. Google Patents. 1996.

  20. Mittal G, Sahana D, Bhardwaj V, Kumar MR. Estradiol loaded PLGA nanoparticles for oral administration: effect of polymer molecular weight and copolymer composition on release behavior in vitro and in vivo. J Control Release. 2007;119(1):77–85.

    Article  CAS  PubMed  Google Scholar 

  21. Mostafa AA, Zaazou MH, Chow LC, Mahmoud AA, Zaki DY, Basha M, et al. Injectable nanoamorphous calcium phosphate based in situ gel systems for the treatment of periapical lesions. Biomed Mater. 2015;10(6):065006.

    Article  PubMed  CAS  Google Scholar 

  22. R Serrano D, H Gallagher K, Marie Healy A. Emerging nanonisation technologies: tailoring crystalline versus amorphous nanomaterials. Curr Top Med Chem. 2015;15(22):2327–40.

    Article  PubMed  CAS  Google Scholar 

  23. Jog R, Kumar S, Shen J, Jugade N, Tan DCT, Gokhale R, et al. Formulation design and evaluation of amorphous ABT-102 nanoparticles. Int J Pharm. 2016;498(1):153–69.

    Article  CAS  PubMed  Google Scholar 

  24. Kumar S, Shen J, Burgess DJ. Nano-amorphous spray dried powder to improve oral bioavailability of itraconazole. J Control Release. 2014;192:95–102.

    Article  CAS  PubMed  Google Scholar 

  25. Yamasaki K, Kwok PCL, Fukushige K, Prud’homme RK, Chan H-K. Enhanced dissolution of inhalable cyclosporine nano-matrix particles with mannitol as matrix former. Int J Pharm. 2011;420(1):34–42.

    Article  CAS  PubMed  Google Scholar 

  26. Abdelwahed W, Degobert G, Stainmesse S, Fessi H. Freeze-drying of nanoparticles: formulation, process and storage considerations. Adv Drug Deliv Rev. 2006;58(15):1688–713.

    Article  CAS  PubMed  Google Scholar 

  27. Bohr A, Water J, Beck-Broichsitter M, Yang M. Nanoembedded microparticles for stabilization and delivery of drug-loaded nanoparticles. Curr Pharm Des. 2015;21(40):5829–44.

    Article  CAS  PubMed  Google Scholar 

  28. Kumar S, Gokhale R, Burgess DJ. Sugars as bulking agents to prevent nano-crystal aggregation during spray or freeze-drying. Int J Pharm. 2014;471(1–2):303–11.

    Article  CAS  PubMed  Google Scholar 

  29. Cerdeira AM, Mazzotti M, Gander B. Formulation and drying of miconazole and itraconazole nanosuspensions. Int J Pharm. 2013;443(1–2):209–20.

    Article  CAS  PubMed  Google Scholar 

  30. Dan J, Ma Y, Yue P, Xie Y, Zheng Q, Hu P, et al. Microcrystalline cellulose-carboxymethyl cellulose sodium as an effective dispersant for drug nanocrystals: a case study. Carbohydr Polym. 2016;136:499–506.

    Article  CAS  PubMed  Google Scholar 

  31. Sofie V, Jan V, Ludo F, Patrick A. Microcrystalline cellulose, a useful alternative for sucrose as a matrix former during freeze-drying of drug nanosuspensions–a case study with itraconazole. Eur J Pharm Biopharm. 2008;70(2):590–6.

    Article  CAS  Google Scholar 

  32. Maniruzzaman M, Nokhodchi A. Continuous manufacturing via hot-melt extrusion and scale up: regulatory matters. Drug Discov Today. 2017;22(2):340–51.

    Article  CAS  PubMed  Google Scholar 

  33. Baumgartner R, Eitzlmayr A, Matsko N, Tetyczka C, Khinast J, Roblegg E. Nano-extrusion: a promising tool for continuous manufacturing of solid nano-formulations. Int J Pharm. 2014;477(1):1–11.

    Article  CAS  PubMed  Google Scholar 

  34. Hao J, Fang X, Zhou Y, Wang J, Guo F, Li F, et al. Development and optimization of solid lipid nanoparticle formulation for ophthalmic delivery of chloramphenicol using a Box-Behnken design. Int J Nanomedicine. 2011;6:683.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Sharma D, Maheshwari D, Philip G, Rana R, Bhatia S, Singh M, et al. Formulation and optimization of polymeric nanoparticles for intranasal delivery of lorazepam using Box-Behnken design: in vitro and in vivo evaluation. Biomed Res Int. 2014;2014:1–14.

    Google Scholar 

  36. Nekkanti V, Muniyappan T, Karatgi P, Hari MS, Marella S, Pillai R. Spray-drying process optimization for manufacture of drug–cyclodextrin complex powder using design of experiments. Drug Dev Ind Pharm. 2009;35(10):1219–29.

    Article  CAS  PubMed  Google Scholar 

  37. Mishra B, Sahoo J, Dixit PK. Enhanced bioavailability of cinnarizine nanosuspensions by particle size engineering: optimization and physicochemical investigations. Mater Sci Eng C. 2016;63:62–9.

    Article  CAS  Google Scholar 

  38. Thakkar HP, Patel BV, Thakkar SP. Development and characterization of nanosuspensions of olmesartan medoxomil for bioavailability enhancement. J Pharm Bioallied Sci. 2011;3(3):426–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhang X, Liu J, Qiao H, Liu H, Ni J, Zhang W, et al. Formulation optimization of dihydroartemisinin nanostructured lipid carrier using response surface methodology. Powder Technol. 2010;197(1–2):120–8.

    Article  CAS  Google Scholar 

  40. Talekar SD, Dave RH. Solubility enhancement of a BCS class II drug using granulated fumed silica as an adsorbent. J Pharm Res 2017;18(6):1–15. https://doi.org/10.9734/JPRI/2017/36872.

    Article  Google Scholar 

  41. Gajera BY, Dugar RP, Dave RH. Formulation development and optimization of ibuprofen poloxamer melt granules using hydrophilic excipients. Br J Pharm Res. 2016;13(6):1–19.

    Article  Google Scholar 

  42. Lu Y, Yu Y, Tang X. Sucrose acetate isobutyrate as an in situ forming system for sustained risperidone release. J Pharm Sci. 2007;96(12):3252–62.

    Article  CAS  PubMed  Google Scholar 

  43. Crowley MM, Zhang F, Repka MA, Thumma S, Upadhye SB, Kumar Battu S, et al. Pharmaceutical applications of hot-melt extrusion: part I. Drug Dev Ind Pharm. 2007;33(9):909–26.

    Article  CAS  PubMed  Google Scholar 

  44. Quek SY, Chok NK, Swedlund P. The physicochemical properties of spray-dried watermelon powders. Chem Eng Process Process Intensif. 2007;46(5):386–92.

    Article  CAS  Google Scholar 

  45. Madgulkar A, Bandivadekar M, Shid T, Rao S. Sugars as solid dispersion carrier to improve solubility and dissolution of the BCS class II drug: clotrimazole. Drug Dev Ind Pharm. 2016;42(1):28–38.

    Article  CAS  PubMed  Google Scholar 

  46. Karolewicz B, Gajda M, Owczarek A, Pluta J, Górniak A. Physicochemical characterization and dissolution studies of solid dispersions of clotrimazole with Pluronic F127. Trop J Pharm Res. 2014;13(8):1225–32.

    Article  CAS  Google Scholar 

  47. Pachuau L, Vanlalfakawma DC, Tripathi SK, Lalhlenmawia H. Muli bamboo (Melocanna baccifera) as a new source of microcrystalline cellulose. 2014.

  48. Mitra A, Kesisoglou F. Impaired drug absorption due to high stomach pH: a review of strategies for mitigation of such effect to enable pharmaceutical product development. Mol Pharm. 2013;10(11):3970–9.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Rutesh H. Dave.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gajera, B.Y., Shah, D.A. & Dave, R.H. Investigating a Novel Hot Melt Extrusion-Based Drying Technique to Solidify an Amorphous Nanosuspension Using Design of Experiment Methodology. AAPS PharmSciTech 19, 3778–3790 (2018). https://doi.org/10.1208/s12249-018-1189-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1208/s12249-018-1189-7

KEY WORDS

Navigation