Skip to main content

Optimizing the Formulation of Poorly Water-Soluble Drugs

  • Chapter
  • First Online:
Formulating Poorly Water Soluble Drugs

Abstract

With as high as 60% of drugs in company pipelines exhibiting poor aqueous solubility, the ability to successfully develop a poorly water-soluble drug has become essential. Gaining a detailed understanding of a compound through preformulation studies can be especially challenging for poorly water-soluble drugs limiting their development. Therefore, this chapter focuses on the application of preformulation studies essential in understanding a poorly water-soluble drug, including solubility studies, solid-state characterization of the active ingredient and formulations thereof, and in vitro and in vivo testing of the lead formulations.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  • Abbas D, Kaloustian J et al (2008) DSC and physico-chemical properties of a substituted pyridoquinoline and its interaction study with excipients. J Therm Anal Calorim 93(2):353–360

    CAS  Google Scholar 

  • Agatonovic-Kustrin S, Markovic N et al (2008) Compatibility studies between mannitol and omeprazole sodium isomers. J Pharm Biomed Anal 48(2):356–360

    PubMed  CAS  Google Scholar 

  • Ahuja S, Rasmussen H (2007) HPLC method development for pharmaceuticals. Elsevier, Boston

    Google Scholar 

  • Ahumada AA, Seeck J, Allemandi D, Manzo RH (1993) The pH/solubility profile of norfloxacin. STP Pharm Sci 3(3):250–253

    CAS  Google Scholar 

  • Alban L, Dahl AK, Hansen AK, Hejgarrd KC, Jensen AL, Kragh M, Thomsen P, Steensgaard P (2001) The welfare impact of increased gavaging doses in rats. Anim Welfare 10(3):303–314

    Google Scholar 

  • AldĂ©n M, Wulff M et al (1995) Influence of selected variables on heat of fusion determinations by oscillating DSC. Thermochim Acta 265:89–102

    Google Scholar 

  • Allen T (1997) Particle size measurement, vol 2, Surface area and pore size determinations. Chapman & Hall, London

    Google Scholar 

  • Alves R, Reis TVdS, Cides da Silva LC, Storpirtis S, Mercuri LP, Matos JdR (2010) Thermal behavior and decomposition kinetics of rifampicin polymorphs under isothermal and non-isothermal conditions. Braz J Pharm Sci 46(2):343–351

    CAS  Google Scholar 

  • Antila R, Yliruusi J (1991) Measurement of specific surface area of pharmaceutical powders by the BET method; effect of drying time and drying temperature. Acta Pharm Nord 3(1):15–18

    CAS  Google Scholar 

  • Avdeef A, Berger CM et al (2000) pH-metric solubility. 2. Correlation between the acid-base titration and the saturation shake-flask solubility-pH methods. Pharm Res 17(1):85–89

    PubMed  CAS  Google Scholar 

  • Baena Y, Barbosa HJ, Jorge A, Martinez F (2004) Estimation of the aqueous solubility of some acetanilide derivatives from octanol-water partition coefficients and entropies of fusion. Acta Farmaceutica Bonaerense 23(1):33–38

    CAS  Google Scholar 

  • Baucells M, Ferrer N, Gomez P, Lacort G, Roura M (1993) Determination of caffeine in solid pharmaceutical samples by FTIR spectroscopy. Mikrochim Acta 112:87–98

    CAS  Google Scholar 

  • Belal F, Elbrashy A, Eid M, Nasr JJ (2008) Stability-indicating HPLC method for the determination of quetiapine: application to tablets and human plasma. J Liq Chromatogr Relat Technol 31:1283–1298

    CAS  Google Scholar 

  • Bergese P, Bontempi E et al (2003) Microstructural investigation of nimesulide-crospovidone composites by X-ray diffraction and thermal analysis. Compos Sci Technol 63(8):1197–1201

    CAS  Google Scholar 

  • Bhalekar MR, Shete TK et al (2010) Solid state photodegradation study of fexofenadine hydrochloride. Anal Lett 43(3):406–416

    CAS  Google Scholar 

  • Bizzi CA, Barin JS, Hermes AL, Mortari SR, Flores EMM (2011) A fast microwave-assisted procedure for loss on drying determination in saccharides. J Braz Chem Soc 22(2):376–381

    CAS  Google Scholar 

  • Bott RF, Oliveira WP (2007) Storage conditions for stability testing of pharmaceuticals in hot and humid regions. Drug Dev Ind Pharm 33:393–401

    PubMed  CAS  Google Scholar 

  • Boyer RF, Simha R (1973) Relation between expansion coefficients and glass temperature: a reply. J Polym Sci Polym Lett Ed 11(1):33–44

    CAS  Google Scholar 

  • Brits M, Liebenberg W, de Villiers MM (2010) Characterization of polymorphic transformations that decrease the stability of tablets containing the WHO essential drug mebendazole. J Pharm Sci 99(3):1138–1151

    PubMed  CAS  Google Scholar 

  • Brown ME (2001) Introduction to thermal analysis. Kluwer, Dordrecht

    Google Scholar 

  • Bruni G, Berbenni V et al (2011) Determination of the nateglinide polymorphic purity through DSC. J Pharm Biomed Anal 54(5):1196–1199

    PubMed  CAS  Google Scholar 

  • Buffeteau T, Desbat B, Eyquem D (1996) Attenuated total reflection fourier transform infrared microspectroscopy: theory and application to polymer samples. Vib Spectrosc 11:29–36

    CAS  Google Scholar 

  • Cameron D, Armstrong EE (1988) Optimization of stepsize in X-ray powder diffractogram collection. Powder Diffr 3(1):32–38

    CAS  Google Scholar 

  • Cammenga HK, Eysel W et al (1993) The temperature calibration of scanning calorimeters: part 2. calibration substances. Thermochim Acta 219:333–342

    CAS  Google Scholar 

  • Campbell Roberts SN, Williams AC et al (2002) Quantitative analysis of mannitol polymorphs. X-ray powder diffractometry–exploring preferred orientation effects. J Pharm Biomed Anal 28(6):1149–1159

    PubMed  CAS  Google Scholar 

  • Cappello B, di Maio C, Iervolino M, Miro A (2007) Combined effect of hydroxypropyl methylcellulose and hydroxypropyl-beta-cyclodextrin on physicochemical and dissolution properties of celecoxib. J Incl Phenom Macrocycl Chem 59:237–244

    CAS  Google Scholar 

  • Carpenter J, Katayama D, Liu L, Chonkaew W, Menard K (2009) Measurement of Tg in lyophilized protein and protein excipient mixtures by dynamic mechanical analysis. J Therm Anal Calorim 95(3):881–884

    CAS  Google Scholar 

  • Chan HK, Ongpipattanakul B, Au-Yeung J (1996) Aggregation of rhDNase occured during the compression of KBr pellets used for FTIR spectroscopy. Pharm Res 13(2):238–242

    PubMed  CAS  Google Scholar 

  • Charsley EL, Laye PG et al (2006) DSC studies on organic melting point temperature standards. Thermochim Acta 446(1–2):29–32

    CAS  Google Scholar 

  • Chawla G, Bansal AK (2008) Improved dissolution of a poorly water soluble drug in solid dispersion with polymeric and non-polymeric hydrophilic additives. Acta Pharm 58:257–274

    PubMed  CAS  Google Scholar 

  • Chitlange SS, Bagri K, Sakarkar DM (2008) Stability indicationg RP-HPLC method for simultaneous ESTIMATION of valsartan and amlodipine in capsule formulation. Asian J Res Chem 1(1):15–18

    CAS  Google Scholar 

  • Condon JB (2006) Surface area and porosity determinations by physisorption: measurements and theory. Elsevier, Amsterdam

    Google Scholar 

  • Corrandini D, Phillips TM (2011) Handbook of HPLC. Taylor & Francis, Boca Raton

    Google Scholar 

  • Craig DQM, Reading M (2007) Thermal analysis of pharmaceuticals. CRC, Boca Raton

    Google Scholar 

  • Cuiping C, Xingrong L, Rujin W (1993) High-performance liquid chromatographic method for the determination of norfloxacin glutamage and glucuronate in solid and liquid dosage forms and its application to stability testing. J Pharm Biomed Anal 11(8):717–721

    Google Scholar 

  • Curatolo W, Nightingale JA, Herbig SM (2009) Utility of hydroxypropylmethylcellulose acetate succinate (HPMCAS) for initiation and maintenance of drug supersaturation in the GI Milieu. Pharm Res 26(6):1419–1431

    PubMed  CAS  Google Scholar 

  • D’Souza AJM, Ford BM et al (2009) Biophysical characterization and formulation of F1–V, a recombinant plague antigen. J Pharm Sci 98(8):2592–2602

    PubMed  Google Scholar 

  • de Villiers MM, Terblanche RJ, Liebenberg W, Swanepoel E, Dekker TG, Song M (2005) Variable-temperature X-ray powder diffraction analysis of the crystal transformation of the pharmaceutically preferred polymorph C of mebendazole. J Pharm Biomed Anal 38:435–441

    PubMed  Google Scholar 

  • Diebold SM, Dressman JB (1998) Dissolved oxygen as a measure for de- and reaeration of aqueous media for dissolution testing. Dissolution Technol 5(3):13–16

    CAS  Google Scholar 

  • Dinnebier RE, Billinge S (2008) Powder diffraction: theory and practice. Royal Society of Chemistry, Cambridge

    Google Scholar 

  • DiNunzio JC, Brough C et al (2010a) Fusion production of solid dispersions containing a heat-sensitive active ingredient by hot melt extrusion and Kinetisol® dispersing. Eur J Pharm Biopharm 74(2):340–351

    PubMed  CAS  Google Scholar 

  • DiNunzio JC, Brough C, Miller DA, Williams ROIII, McGinity JW (2010b) Applications of KinetiSol dispersing for the production of plasticizer free amorphous solid dispersions. Eur J Pharm Sci 40(3):179–187

    PubMed  CAS  Google Scholar 

  • DiNunzio JC, Hughey JR, Brough C, Miller DA, Williams RO III, McGinity JW (2010c) Production of advanced solid diserpsions for enhanced bioavailability of itraconazole using kinetisol dispersing. Drug Dev Ind Pharm 36(9):1064–1078

    PubMed  CAS  Google Scholar 

  • DiNunzio JC, Miller DA, Yang W, McGinity JW, Williams RO III (2008) Amorphous compositions using concentration enhancing polymers for improved bioavailability of itraconazole. Mol Pharm 5(6):968–980

    PubMed  CAS  Google Scholar 

  • Dios Lopez-Gonzalez J, Carpenter FG, Deitz VR (1955) Adsorption of nitrogen on carbon adsorbents at low pressures between 69 and 90°K. J Res Natl Bur Stand (US) 55:11–18

    Google Scholar 

  • Duddu SP, Weller K (1996) Importance of glass transition temperature in accelerated stability testing of amorphous solids: case study using a lyophilized aspirin formulation. J Pharm Sci 85(3):345–347

    PubMed  CAS  Google Scholar 

  • Dwivedi AM (2002) Residual solvent analysis in pharmaceuticals. Pharm Technol November: 42–46

    Google Scholar 

  • Engstrom JD, Simpson DT, Lai ES, Williams RO III, Johnston KP (2007) Morphology of protein particles produced by spray freezing of concentrated solutions. Eur J Pharm Biopharm 64:149

    Google Scholar 

  • Fagerlund G (1973) Determination of specific surface by the BET method. Materiaux et Constructions 6(33):239–245

    CAS  Google Scholar 

  • Fahr A, Liu X (2007) Drug delivery strategies for poorly water-soluble drugs. Expert Opin Drug Deliv 4(4):403–416

    PubMed  CAS  Google Scholar 

  • Ferrari PC, Oliveira GF, Chibebe FCS, Evangelista RC (2009) In vitro characterization of coevaporates containing chitosan for colonic drug delivery. Carbohydr Polym 78:557–563

    CAS  Google Scholar 

  • Filho R, Franco P et al (2009) Stability studies on nifedipine tablets using thermogravimetry and differential scanning calorimetry. J Therm Anal Calorim 97(1):343–347

    CAS  Google Scholar 

  • Fortunato D (2005) Dissolution method development for immediate release solid oral dosage forms. Dissolution Technol August:12–14, August

    Google Scholar 

  • Fotaki N, Symillides M et al (2005) Canine versus in vitro data for predicting input profiles of l-sulpiride after oral administration. Eur J Pharm Sci 26(3–4):324–333

    PubMed  CAS  Google Scholar 

  • Friesen DT, Shanker R, Crew M, Smithey DT, Curatolo WJ, Nightingale JAS (2008) Hydroxypropyl methylcellulose acetate succinate-based spray-dried dispersions: an overview. Mol Pharm 5(6):1003–1019

    PubMed  CAS  Google Scholar 

  • Gad SC (2008) Pharmaceutical manufacturing handbook: regulations and quality. Wiley, Hoboken

    Google Scholar 

  • Gao Z, Moore TW et al (2006) Effects of deaeration methods on dissolution testing in aqueous media: a study using a total dissolved gas pressure meter. J Pharm Sci 95(7):1606–1613

    PubMed  CAS  Google Scholar 

  • Gordon M, Taylor JS (1952) Ideal copolymers and the second-order transitions of synthetic rubbers. I. noncrystalline copolymers. J Appl Chem 2:493–500

    CAS  Google Scholar 

  • Goskonda VR, Reddy IK, Durrant MJ, Wilber W, Khan MA (1998) Solid-state stability assessment of controlled release tablets containing Carbopol 971P. J Control Release 54:87–93

    PubMed  CAS  Google Scholar 

  • Gregg SJ, Sing KSW (1982) Adsorption surface area and porosity. Academic, New York

    Google Scholar 

  • Hartauer KJ, Miller ES et al (1992) Diffuse reflectance infrared fourier transform spectroscopy for the quantitative analysis of mixtures of polymorphs. Int J Pharm 85(1–3):163–174

    CAS  Google Scholar 

  • Heigoldt U, Sommer F et al (2010) Predicting in vivo absorption behavior of oral modified release dosage forms containing pH-dependent poorly soluble drugs using a novel pH-adjusted biphasic in vitro dissolution test. Eur J Pharm Biopharm 76(1):105–111

    PubMed  CAS  Google Scholar 

  • Heng D, Cutler D et al (2008) What is a suitable dissolution method for drug nanoparticles? Pharm Res 25(7):1696–1701

    PubMed  CAS  Google Scholar 

  • Hughey JR, DiNunzio JC, Bennett RC, Brough C, Miller DA, Hua M, Williams RO III, McGinity JW (2010) Dissolution enhancement of a drug exhibiting thermal and acidic decomposition characteristics by fusion processing: a comparative study to hot melt extrusion and kinetisol dispersing. AAPS PharmSciTech 11(2):760–774

    PubMed  CAS  Google Scholar 

  • Igwe GJI (1991) Powder technology and multiphase systems: gas permeametry and surface area measurement. Ellis Horwood Limited, Chichester

    Google Scholar 

  • Jain N, Yalkowsky SH (2001) Estimation of the aqueous solubility I: application to organic nonelectrolytes. J Pharm Sci 90(2):234–252

    PubMed  CAS  Google Scholar 

  • Jantratid E, Janssen N et al (2008) Dissolution media simulating conditions in the proximal human gastrointestinal tract: an update. Pharm Res 25(7):1663–1676

    PubMed  CAS  Google Scholar 

  • Joshi BV, Patil VB, Pokharkar VB (2002) Compatibility studies between carbamazepine and tablet excipients using thermal and non-thermal methods. Drug Dev Ind Pharm 28(6):687–694

    PubMed  CAS  Google Scholar 

  • Joy AS (1953) Methods and techniques for the determination of specific surface by gas adsorption. Vacuum 3:254

    Google Scholar 

  • Juenemann D, Jantratid E et al (2011) Biorelevant in vitro dissolution testing of products containing micronized or nanosized fenofibrate with a view to predicting plasma profiles. Eur J Pharm Biopharm 77(2):257–264

    PubMed  CAS  Google Scholar 

  • Junghanns JAH, Mueller RH (2008) Nanocrystal technology, drug delivery and clinical applications. Int J Nanomed 3(3):295–309

    CAS  Google Scholar 

  • Khan MA, Karnachi AA, Agarwal V, Vaithiyalingam SR, Nazzal S, Reddy IK (2000) Stability characterization of controlled release coprecipitates and solid dispersions. J Control Release 63:1–6

    PubMed  CAS  Google Scholar 

  • Khomane K, Kumar L et al (2011) NP-647, a novel TRH analogue: investigating physicochemical parameters critical for its oral and parenteral delivery. Int J Pharm 406(1–2):21–30

    PubMed  CAS  Google Scholar 

  • Kimura N, Fukui H, Takagaki H, Yonemochi E, Terada K (2001) Characterization of polymorphs of a novel quinolinone derivative, TA-270 (4-Hydroxy-1-methyl-3-octyloxy-7-sinapinoylamino-2(1 H)-quinolinone). Chem Pharm Bull 49(10):1321–1325

    PubMed  CAS  Google Scholar 

  • Kirk JH, Blatchford CG (2007) Lactose: a definitive guide to polymorph determination. Int J Pharm 334:103–114

    PubMed  CAS  Google Scholar 

  • Konno H, Handa T, Alonzo DE, Taylor LS (2008) Effect of polymer type on the dissolution profile of amorphous solid dispersions containing felodipine. Eur J Pharm Biopharm 70:493–499

    PubMed  CAS  Google Scholar 

  • Lakshman JP, Cao Y, Kowalski J, Serajuddin ATM (2008) Application of melt extrusion in the development of a physically and chemically stable high-energy amorphous solid dispersion of a poorly water-soluble drug. Mol Pharm 5(6):994–1002

    PubMed  CAS  Google Scholar 

  • Lee EH, Smith DT, Fanwick PE, Byrn SR (2010) Characterization and anisotropic lattice expansion/contraction of polymorphs of tenofovir disoproxil fumarate. Crys Growth Des 10:2314–2322

    CAS  Google Scholar 

  • Lee HG, Zhang GGZ et al (2011) Cocrystal intrinsic dissolution behavior using a rotating disk. J Pharm Sci 100(5):1736–1744

    PubMed  CAS  Google Scholar 

  • Lenhardt T, Vergnault G, Grenier P, Scherer D, Langguth P (2008) Evaluation of nanosuspensions for absorption enhancement of poorly soluble drugs: in vitro transport studies across intestinal epithelial monolayers. AAPS J 10(3):435–438

    PubMed  CAS  Google Scholar 

  • Li S, Wong S et al (2005) Investigation of solubility and dissolution of a free base and two different salt forms as a function of pH. Pharm Res 22(4):628–635

    PubMed  CAS  Google Scholar 

  • Liu F-h, Jiang Y (2007) Room temperature ionic liquid as matrix medium for the determination of residual solvents in pharmaceuticals by static headspace gas chromatography. J Chromatogr A 1167(1):116–119

    PubMed  CAS  Google Scholar 

  • Liu XB, Ye JX, Quan LH, Liu CY, Deng XL, Yang M, Liao YH (2008) Pulmonary delivery of scutellarin solution and mucoadhesive particles in rats. Eur J Pharm Biopharm 70:845–852

    PubMed  CAS  Google Scholar 

  • Lowell S, Shields JE (1991) Powder surface area and porosity. powder technology series. b. scarlett. Chapman & Hall, New York, pp 155–173

    Google Scholar 

  • Martins MT, Paim CS et al (2010) Development of a dissolution test for lamotrigine in tablet form using an ultraviolet method. Braz J Pharm Sci 46:179–186

    CAS  Google Scholar 

  • Mashru RC, Sutariya VB et al (2005) Characterization of solid dispersions of rofecoxib using differential scanning calorimeter. J Therm Anal Calorim 82(1):167–170

    CAS  Google Scholar 

  • McAllister M (2010) Dynamic dissolution: a step closer to predictive dissolution testing? Mol Pharm 7(5):1374–1387

    CAS  Google Scholar 

  • McConville JT, Overhoff KA, Sinswat P, Vaughn JM, Frei BL, Burgess DS, Talbert RL, Peters JI, Johnston KP, Williams RO III (2006) Targeted high lung concentrations of itraconazole using nebulized dispersions in a murine model. Pharm Res 23(5):901–911

    PubMed  CAS  Google Scholar 

  • McConville JT, Williams RO III, Carvalho TC, Iberg AN, Johnston KP, Talbert RL, Burgess D, Peters JI (2005) Design and evaluation of a restraint-free small animal inhalation dosing chamber. Drug Dev Ind Pharm 1:35–42

    Google Scholar 

  • Menegola J, Steppe M et al (2007) Dissolution test for citalopram in tablets and comparison of in vitro dissolution profiles. Eur J Pharm Biopharm 67(2):524–530

    PubMed  CAS  Google Scholar 

  • Miller DA, DiNunzio JC, Yang W, McGinity JW, Williams RO III (2008) Enhanced in vivo absorption of itraconazole via stabilization of supersaturation followign acidic-to-neutral pH transition. Drug Dev Ind Pharm 34:890–902

    PubMed  CAS  Google Scholar 

  • Mora PC, Cirri M et al (2006) Differential scanning calorimetry as a screening technique in compatibility studies of DHEA extended release formulations. J Pharm Biomed Anal 42(1):3–10

    PubMed  CAS  Google Scholar 

  • Morello M, Krone CL, Dickerson S, Howerth E, Germishuizen WA, Wong YL, Edwards D, Bloom BR, Hondalus MK (2009) Dry-powder pulmonary insufflation in the moues for application to vaccine or drug studies. Tuberculosis 89:371–377

    PubMed  CAS  Google Scholar 

  • Mosharraf M, Sebhatu T, Nystrom C (1999) The effects of disordered structure on the solubility and dissolution rates of some hydrophilic, sparingly soluble drugs. Int J Pharm 177:29–51

    PubMed  CAS  Google Scholar 

  • Mulligan KJ, McCauley H (1995) Factors that influence the determination of residual solvents in pharmaceuticals by automated static headspace sampling coupled to capillary GCMS. J Chromatogr Sci 33:49–54

    PubMed  CAS  Google Scholar 

  • Mura P, Bettinetti GP et al (1998a) Differential scanning calorimetry in compatibility testing of picotamide with pharmaceutical excipients. Thermochim Acta 321(1–2):59–65

    CAS  Google Scholar 

  • Mura P, Faucci MT et al (1998b) Compatibility study between ibuproxam and pharmaceutical excipients using differential scanning calorimetry, hot-stage microscopy and scanning electron microscopy. J Pharm Biomed Anal 18(1–2):151–163

    PubMed  CAS  Google Scholar 

  • O’Donnell KP, Williams RO III (2011) Nanoparticulate systems for oral drug delivery to the colon. Int J Nanotechnol 8(1/2):4–20

    Google Scholar 

  • Okazaki A, Mano T, Sugano K (2008) Theoretical dissolution model of poly-disperse drug particles in biorelevant media. J Pharm Sci 97(5):1843–1852

    PubMed  CAS  Google Scholar 

  • Overhoff KA, McConville JT, Yang W, Johnston KP, Peters JI, Williams RO III (2008) Effect of stabilizer on the maximum degree and extent of supersaturation and oral absorption of tacrolimus made by ultra-rapid freezing. Pharm Res 25(1):167–175

    PubMed  CAS  Google Scholar 

  • Overhoff KA, Moreno A, Miller DA, Johnston KP, Williams RO III (2007) Solid dispersions of itraconazole and enteric polymers made by ultra-rapid freezing. Int J Pharm 336:122–132

    PubMed  CAS  Google Scholar 

  • Paim CS, Martins MT, Malesuik MD, Steppe M (2010) LC determination of entacapone in tablets: in vitro dissolution studies. J Chromatogr Sci 48:755–759

    PubMed  CAS  Google Scholar 

  • Patton JS (1996) Mechanisms of macromolecule absorption by the lungs. Adv Drug Deliv Rev 19:3–36

    CAS  Google Scholar 

  • Patton JS, Byron PR (2007) Inhaling medicines: delivering drugs to the body through the lungs. Nat Rev 6:67–74

    CAS  Google Scholar 

  • Pavon JLP, del Nogal Sanchez M, Pinto CG, Laespada MEF, Cordero BM (2006) Use of mass spectrometry methods as a strategy for detection and determination of residual solvents in pharmaceutical products. Anal Chem 78:4901–4908

    Google Scholar 

  • Pendharkar CM, Jhawar RJ, Rutledge JM, Hause W, Grim WM, Harwood RJ (1990) Influence of the specific surface area of selected raw materials on the granulation process using an instrumented mixer. Pharmaceut Tech 14(4):44–53

    CAS  Google Scholar 

  • Phalen RF, Mannix RC, Drew RT (1984) Inhalation exposure methodology. Environ Health Perspect 56:23–34

    PubMed  CAS  Google Scholar 

  • Qian F, Huang J, Zhu Q, Haddadin R, Gawel J, Garmise R, Hussain M (2010) Is a distinctive single Tg a reliable indicator for the homogeneity of amorphous solid dispersion? Int J Pharm 395:232–235

    PubMed  CAS  Google Scholar 

  • Radha S, Gutch PK et al (2010) Thermal analysis of interactions between an oxime and excipients in some binary mixtures by differential scanning calorimetry and thermagravimetric analysis. J Pharm Res 3(3):590–595

    Google Scholar 

  • Ran Y, He Y et al (2002) Estimation of aqueous solubility of organic compounds by using the general solubility equation. Chemosphere 48(5):487–509

    PubMed  CAS  Google Scholar 

  • Rogers TL, Nelsen AC, Hu J, Brown JN, Sarkari M, Young TJ, Johnston KP, Williams RO III (2002) A novel particle engineering technology to enhance dissolution of poorly water soluble drugs: spray-freezing into liquid. Eur J Pharm Biopharm 54:271–280

    PubMed  CAS  Google Scholar 

  • Roy CJ, Hale M, Hartings JM, Pitt L, Duniho S (2003) Impact of inhalation exposure modality and particle size on the respiratory deposition of ricin in BALB/c mice. Inhal Toxicol 15(6):619–638

    PubMed  CAS  Google Scholar 

  • Salari A, Young RE (1998) Application of attenuated total reflectance FTIR spectroscopy to the analysis of mixtures of pharmaceutical polymorphs. Int J Pharm 163:157–166

    CAS  Google Scholar 

  • Sandell E (1993) Industrial aspects of pharmaceutics. Swedish Pharmaceutical, Stockholm

    Google Scholar 

  • Sauer BB, Kampert WG et al (2000) Temperature modulated DSC studies of melting and recrystallization in polymers exhibiting multiple endotherms. Polymer 41(3):1099–1108

    CAS  Google Scholar 

  • Schawe JEK (1996) Modulated temperature DSC measurements: the influence of the experimental conditions. Thermochim Acta 271:127–140

    CAS  Google Scholar 

  • Schick C (2009) Differential scanning calorimetry (DSC) of semicrystalline polymers. Anal Bioanal Chem 395(6):1589–1611

    PubMed  CAS  Google Scholar 

  • Seedher N, Kanojia M (2009) Co-solvent solubilization of some poorly-soluble antidiabetic drugs. Pharm Dev Technol 14(2):185–192

    PubMed  CAS  Google Scholar 

  • Shaw LR, Irwin WJ et al (2005) The effect of selected water-soluble excipients on the dissolution of paracetamol and ibuprofen. Drug Dev Ind Pharm 31(6):515–525

    PubMed  CAS  Google Scholar 

  • Shi Y, Gao P et al (2010) Application of a biphasic test for characterization of in vitro drug release of immediate release formulations of celecoxib and its relevance to in vivo absorption. Mol Pharm 7(5):1458–1465

    CAS  Google Scholar 

  • Sing KSW, Everett DH, Haul RAW, Moscou L, Pierotti RA, Rouquerol J, Siemieniewska T (1985) Reporting physisorption data for gas/solid systems with special reference to the determination of surface area and porosity. Pure Appl Chem 57(4):603–619

    CAS  Google Scholar 

  • Singh S, Singh B et al (2006) Stress degradation studies on ezetimibe and development of a validated stability-indicating HPLC assay. J Pharm Biomed Anal 41(3):1037–1040

    PubMed  CAS  Google Scholar 

  • Sitar Curin A, Greman M, Vrecer F, Kotar-Jordan B, Sustar B (1997) Study of crystal modifications of lansoprazole using FT-IR spectroscopy, solid-state NMR spectroscopy and FT-Raman spectroscopy. Farm Vestn 48:290–291

    Google Scholar 

  • Stanisz B, Kania L (2006) Validation of HPLC Method for determination of atorvastatin in tablets and for monitoring stability in solid phase. Acta Poloniae Pharmaceutica - Drug Research 63(6):471–476

    CAS  Google Scholar 

  • Stark G, Fawcett JP, Tucker IG, Weatherall IL (1996) instrumental evaluation of color of solid dosage forms during stability testing. Int J Pharm 143:93–100

    CAS  Google Scholar 

  • Sung JC, Padilla DJ, Garcia-Contreras L, VerBerkmoes JL, Durbin D, Peloquin CA, Elbert KJ, Hickey AJ, Edwards DA (2009) Formulation and pharmacokinetics of self-assembled rifampicin nanoparticle systems for pulmonary delivery. Pharm Res 26(8):1847–1855

    PubMed  CAS  Google Scholar 

  • Surendrakumar K, Martyn GP, Hodgers ECM, Jansen M, Blair JA (2003) Sustained release of insulin from sodium hyaluronate based dry powder formulation after pulmonary delivery in beagle dogs. J Control Release 91:385–394

    PubMed  CAS  Google Scholar 

  • Swinkels D, Bristow N, Gale L (1994) Effect of sample preparation on the BET surface area of EMD. Progress in Batteries & Battery Materials, Belgium

    Google Scholar 

  • Syam P, Sundaramoorthy K, Vetrichel VT (2010) Design of biodegradable polymer nanoparticles for oral drug delivery of stavudine: in vitro dissolution studies and characterization. Int J Pharm Technol 3(1):1360–1372

    Google Scholar 

  • Teijeiro SA, BriĂ±Ă³n MC (2006) 3′-azido-3′-deoxy-5′-o-isonicotinoylthymidine: a novel antiretroviral analog of zidovudine. III. Solubility studies. Nucleosides Nucleotides Nucleic Acids 25(2):191–202

    PubMed  CAS  Google Scholar 

  • Tho I, Liepold B et al (2010) Formation of nano/micro-dispersions with improved dissolution properties upon dispersion of ritonavir melt extrudate in aqueous media. Eur J Pharm Sci 40(1):25–32

    PubMed  CAS  Google Scholar 

  • Tiwari M, Chawla G et al (2007) Quantification of olanzapine polymorphs using powder X-ray diffraction technique. J Pharm Biomed Anal 43(3):865–872

    PubMed  CAS  Google Scholar 

  • Tobyn M, Brown J et al (2009) Amorphous drug–PVP dispersions: application of theoretical, thermal and spectroscopic analytical techniques to the study of a molecule with intermolecular bonds in both the crystalline and pure amorphous state. J Pharm Sci 98(9):3456–3468

    PubMed  CAS  Google Scholar 

  • Tomoda K, Ohkoshi T, Hirota K, Sonavane GS, Nakajima T, Terada H, Komuro M, Kitazato K, Makino K (2009) Preparation and properties of inhalable nanocomposite particles for treatment of lung cancer. Colloids Surf B Biointerfaces 71:177–182

    PubMed  CAS  Google Scholar 

  • Vandecruys R, Peeters J, Verreck G, Brewster ME (2007) Use of a screening method to determine excipients which optimize the extent and stability of supersaturated drug solutions and application of this system to solid formulation design. Int J Pharm 342:168–175

    PubMed  CAS  Google Scholar 

  • Venkatesh S, Li J et al (1996) Intrinsic solubility estimation and pH-solubility behavior of cosalane (NSC 658586), an extremely hydrophobic diprotic acid. Pharm Res 13(10):1453–1459

    PubMed  CAS  Google Scholar 

  • Vertzoni M, Dressman J, Butler J, Hempenstall J, Reppas C (2005) Simulation of fasting gastric conditions and its importance for the in vivo dissolution of lipophilic compounds. Eur J Pharm Biopharm 60:413–417

    PubMed  CAS  Google Scholar 

  • Viegas TX, Curatella RU, Van Winkle LL, Brinker G (2001a) Intrinsic drug dissolution testing using the stationary disk system. Dissolution Technol 8(3):19–23

    CAS  Google Scholar 

  • Viegas TX, Curatella RU, Van Winkle LL, Brinker G (2001b) Measurement of intrinsic drug dissolution rates using two types of apparatus. Pharm Technol 25(6):44–53

    CAS  Google Scholar 

  • Wang J, Davidovich M et al (2010) Solid-state interactions of a drug substance and excipients and their impact on tablet dissolution: a thermal–mechanical facilitated process-induced transformation or PIT. J Pharm Sci 99(9):3849–3862

    PubMed  CAS  Google Scholar 

  • Wang Y, Chow MSS et al (2008) Mechanistic analysis of pH-dependent solubility and trans-membrane permeability of amphoteric compounds: application to sildenafil. Int J Pharm 352(1–2):217–224

    PubMed  CAS  Google Scholar 

  • Waterman KC, Adami RC (2005) Accelerated aging: prediction of chemical stability of pharmaceuticals. Int J Pharm 293(1–2):101–125

    PubMed  CAS  Google Scholar 

  • Webb PA, Orr C (1997) Analytical methods in fine particle technology. Micromeritics Instrument Corporation, Norcross

    Google Scholar 

  • Wempe MF, Buchanan CM et al (2007) Pharmacokinetics of letrozole in male and female rats: influence of complexation with hydroxybutenyl-β-cyclodextrin. J Pharm Pharmacol 59(6):795–802

    PubMed  CAS  Google Scholar 

  • Williams AC, Cooper VB, Thomas L, Griffith LJ, Petts CR, Booth SW (2004) Evaluation of drug physical form during granulation, tabletting and storage. Int J Pharm 275:29–39

    PubMed  CAS  Google Scholar 

  • Witschi C, Doelker E (1997) Residual solvents in pharmaceutical products: acceptable limits, influences on physicochemical properties, analytical methods and documented values. Eur J Pharm Biopharm 43(3):215–242

    CAS  Google Scholar 

  • Wolkers WF, Oldenhof H (2005) In situ FTIR assessment of dried Lactobacillus bulgaricus: KBr disk formation affects physical properties. Spectroscopy 19:89–99

    CAS  Google Scholar 

  • Xia D, Cui F, Piao H, Cun D, Piao H, Jiang Y, Ouyang M, Quan P (2010) Effect of crystal size on the In Vitro dissolution and oral absorption of nitrendipine in rats. Pharm Res 27:1965–1976

    PubMed  CAS  Google Scholar 

  • Yalkowsky SH, Valvani SC (1980) Solubility and partitioning I: solubility of nonelectrolytes in water. J Pharm Sci 69(8):912–922

    PubMed  CAS  Google Scholar 

  • Yang L, Wu X et al (2009) Novel biodegradable polylactide/poly(ethylene glycol) micelles prepared by direct dissolution method for controlled delivery of anticancer drugs. Pharm Res 26(10):2332–2342

    PubMed  CAS  Google Scholar 

  • Yang W, Johnston KP, Williams RO II (2010) Comparison of bioavailability of amorphous versus crystalline itraconazole nanoparticles via pulmonary administration in rats. Eur J Pharm Biopharm 75:33–41

    PubMed  CAS  Google Scholar 

  • Yang W, Tam J, Miller DA, Zhou J, McConville JT, Johnston KP, Williams RO III (2008) High bioavailability from nebulized itraconazole nanoparticle dispersions with biocompatible stabilizers. Int J Pharm 361:177–188

    PubMed  CAS  Google Scholar 

  • Yu LX, Carlin AS et al (2004) Feasibility studies of utilizing disk intrinsic dissolution rate to classify drugs. Int J Pharm 270(1–2):221–227

    PubMed  CAS  Google Scholar 

  • Zakeri-Milani P, Barzegar-Jalali M, Azimi M, Valizadeh H (2009) Biopharmaceutical classification of drugs using intrinsic dissolution rate (IDR) and rat intestinal permeability. Eur J Pharm Biopharm 73:102–106

    PubMed  CAS  Google Scholar 

  • Zhao C, Furukawa S, Ohki Y (2009) A novel collagenase-assisted extraction of active pharmaceutical ingredients from gelatin products for quantitative analysis by high performance liquid chromatography. J Chromatogr A 1216:4524–4528

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Robert O. Williams III .

Editor information

Editors and Affiliations

Appendices

Method Capsule 1pH-Solubility Profile by Direct Determination in Aqueous Suspension

Based on the method reported by Li et al. (2005)

Objective

  • To determine the pH-solubility profile of haloperidol and the corresponding hydrochloride and mesylate salts.

Equipment and Reagents

  • Haloperidol free base

  • Haloperidol hydrochloride

  • Haloperidol mesylate

  • Deionized water

  • Hydrochloric acid solution

  • Sodium hydroxide solution

  • 10 mL sealable vials

  • Water bath or environmental shaker capable of maintaining 37°C

  • 0.45 μm acrodisc filters w/attached syringe

  • UV spectrophotometer (250 nm detection wavelength)

Method

  • Add 5 mL of water to each 10 mL vial, 9 total, 3 for each compound.

  • Place excess solids (one compound per vial) in the prefilled vials such that a suspension results.

  • Titrated each vial to pH 1 via HCl and NaOH solution addition.

  • Equilibrate the vials at 37°C for 24 h. Note: agitation recommended during equilibration if possible.

  • Following equilibration, verify that no pH shift has occurred.

  • Remove an aliquot of the suspension and filter through a 0.45 μm (or smaller) filter.

  • Dilute sample with suitable organic solvent (i.e., acetonitrile) to obtain concentrations in the working linear range of the spectrophotometer.

  • Analyze samples on UV spectrophotometer at 250 nm.

  • Titrate each vial with NaOH and HCl solutions to pH 2.

  • Equilibrate vials for 24 h under identical conditions.

  • Confirm no pH shift upon equilibration.

  • Repeat sampling and analysis procedure as above.

  • Continue titration, equilibration, sampling procedure through pH range of 1–13, or desired regions thereof.

Results

  • Plotting the solubility (ug/mL) versus pH of the free base and its HCl salt revealed a significant drop in solubility of both compounds above pH 5. Additionally, the plot demonstrates no significant difference in the aqueous solubility between the free base and HCl salt through pH 7.

  • The pH versus solubility plot for the mesylate salt of haloperidol revealed significantly higher solubilities of the API through pH 5. Similar to the free base and HCl salt, solubility of the API decreased significantly above pH 5.

Method Capsule 2Analysis of Drug–Excipient Interactions Via Differential Scanning Calorimetry

Based on the method reported by Mura et al. (1998a) Thermochimica Acta.

Objective

  • To determine compatibility of picotamide in the presence of excipients commonly used in tableting formulations.

Equipment and Reagents

  • Picotamide recrystallized from water–ethanol 8:1.

  • Excipients: PVP K30, PVPXL, tartaric acid, ascorbic acid, hydroxypropylmethyl cellulose, hydroxyethyl cellulose, sodium carboxymethyl cellulose, microcrystalline cellulose, Veegum F, Arabic gum, cornstarch.

  • Mortar pestle

  • Microbalance (mg scale)

  • Differential scanning calorimeter

  • Aluminum pans with perforated lits

Method

  • Sieve each material and obtain the 75–150 μm fraction for analysis.

  • Prepare individual physical mixtures the API and each excipient (1:1) in a mortar using a spatula to gently blend the components.

  • Prepare co-ground mixtures of each drug:excipient combination by grinding an aliquot of the corresponding physical mixture in a mortar with a pestle for 10 min.

  • Prepare kneaded mixtures of each drug:excipient combination by slurring an aliquot of the corresponding physical mixture with ethanol (1–2 mL) and grinding in a mortar with pestle to obtain a paste. Dry under vacuum in a desiccator at room temperature to a constant weight.

  • Place aliquots of physical mixtures at 60°C for drug–excipient storage stability analysis.

  • Analyze each individual component, physical mixture, co-ground mixture, kneaded mixture, and stability sample in the following manner: Place an aluminum pan (without lid) on the microbalance and zero the scale. Fill the pan with approximately 5–10 mg of the material for analysis. Place the filled pan back on the balance and record the exact weight of filling. Place a lid in the filled pan. Place the pan in the DSC apparatus on the sample side. A reference pan with lid should be placed on the reference cell. Perform scan at 10 K/min from 30–200°C.

Results

  • Picotamide monohydrate exhibits an endothermic event at 123.0  ±  2.4°C.

  • Maintenance of the anhydrous state for physical mixtures and co-ground mixtures with microcrystalline cellulose, cornstarch, methocel, and ethocel indicate compatibility.

  • Hydration of the API in physical mixtures with Veegum and Arabic gum indicates incompatibility.

  • Fresh and stored physical blends with PVP exhibited no interactions. The presence of the dehydration peak at 124°C for ground mixtures with PVP-XL indicates incompatibility. The absence of the picotamide melting endotherm for co-ground mixtures with PVP-K30 indicates amorphization of the API and dissolution into the polymeric carrier.

  • The presence of acidic excipients yielded broadening and downshift of thermal effects followed by exothermic decomposition for all samples indicating strong incompatibility.

Method Capsule 3X-Ray Diffraction Parameter Optimization

Based on the method reported by Tiwari et al. (2007)

Objective

  • To optimize the scan parameters for X-ray diffraction analysis including step size and dwell time as well as to assess the influence of particle size.

Equipment and Reagents

  • Olanzapine polymorphs I and II

  • X-ray diffractometer

  • Poly methyl methacrylate sample holder or equivalent

  • Glass microscope slide or similar

Method

  • Pass polymorph I through multiple sieves (i.e., BSS #80, 120, and 240) collecting aliquots of each sieve fraction.

  • Optimization of scan rate–scan a 5% w/w mixture of polymorph I in polymorph II over the range of 3–40° 2Q under the following conditions and identify the parameters capable of providing the greatest number of identified peaks in the least amount of time:

  • Step time of 0.5 s, step size of 0.025°

  • Step time of 0.5 s, step size of 0.0125°

  • Step time of 1 s, step size of 0.0125°

  • Step time of 5 s, step size of 0.05°

  • Step time of 5 s, step size of 0.0125°

  • Using the selected optimized scan rate, analyze the aliquots of each sieve fraction to assess the influence of particle size.

Results

  • A step time of 5 s and step sizes of 0.05 and 0.0125° allowed identification of four distinct peaks.

  • Lower step times of 1 s and 0.5 s allowed identification of only two and one peaks, respectively, regardless of step size.

  • The step size of 0.05° was selected as it drastically reduced the scan time from 246.66 min to 61.66 min.

  • Particle size significantly influenced the number of identifiable peaks.

  • Sieve fraction BSS #120/240 significantly improved resolution compared to larger particle-size fractions.

Method Capsule 4Accelerated Stability Monitoring of Amorphous Solid Dispersions

Based on the method reported by DiNunzio et al. (2010a, b, c)

Objective

  • To assess formulation stability against recrystallization upon storage for amorphous itraconazole compositions produced by hot–melt extrusion and Kinetisol dispersing.

Equipment and Reagents

  • High-density polyethylene (HDPE) bottles or similar with induction sealing capability.

  • Oven capable of maintaining 40°C  ±  1°C.

  • Saturated salt solution capable of maintaining 75% relative humidity within 40°C oven.

  • X-ray diffractometer (XRD)

  • Sample holder for XRD

Method

  • Analyze bulk itraconazole and excipients individually by XRD over the scan range of 5–50° 2Θ with a step size of 0.05° and a dwell time of 3 s. Identify major characteristic peaks to be used in subsequent analysis.

  • Analyze physical mixtures of drug and excipients at ratios identical to those used for the final formulation by XRD employing the same parameters as above. Identify the major characteristic peaks of itraconazole present in physical mixtures.

  • Following formulation production, immediately obtain XRD profiles for each product using identical scan parameters as before. Identify major characteristic peaks of itraconazole and corresponding intensities if present.

  • Place 2 g of a single formulation into a 30-mL HDPE bottle.

  • Prepare three bottles for each formulation for each of the three time points.

  • Induction seal each bottle.

  • Verify induction seal robustness prior to placing on stability.

  • Place all samples in 40°C 75% RH oven.

  • At 1 month remove three samples of each formulation for analysis.

  • Allow samples to equilibrate to room temperature for 24 h.

  • Open containers and analyze powders individually by XRD using identical parameters as described above for characteristic crystalline peaks of itraconazole.

  • Repeat sample removal, equilibration, and analysis at 3 and 6 months.

  • Generate a plot for the XRD data of intensity versus angle (degrees 2Θ) for all formulations at a single time point for comparison. Do this for each time point.

Results

  • Analysis of bulk itraconazole revealed numerous characteristic crystalline peaks between 10 and 35° 2Θ.

  • XRD diffraction patterns of formulations immediately post production exhibit amorphous halos and lack any characteristic itraconazole peaks.

  • Materials produced by kinetisol dispersing which contained no plasticizer exhibited no peak growth over time when stored at accelerated conditions.

  • Formulations produced by hot–melt extrusion exhibited gradual growth of characteristic itraconazole peaks, indicating recrystallization upon storage.

Method Capsule 5BET Specific Surface Area Determination for a High Surface-Area Heat-Liable Material

Based on the method reported by Engstrom et al. (2007)

Objective

  • To determine the specific surface area of protein powders produced by spray freezing into liquid.

Equipment and Reagents

  • Protein powder produced by spray freezing into liquid.

  • Quantachrome Nova 2000 BET apparatus including sample cells.

  • Dry box

  • Liquid nitrogen

  • Nitrogen gas: high purity, dry

  • Analytical balance

Method

  • Using the analytical balance, weigh the empty sample cells and record the weights.

  • Within the dry box, add powder sample to the BET sample cells. As the device has two cells which can be analyzed simultaneously, all powders for analysis are to be analyzed in duplicate.

  • Using the analytical balance record the weight of the full sample cell.

  • Attach filled sample cells to degassing station ports.

  • Engage vacuum

  • Allow samples to degas under vacuum for 12 h.

  • Fill liquid nitrogen Dewar with liquid nitrogen to the maximum fill level.

  • Repressurize the system, remove sample cells, and, using the analytical balance, immediately record the weights of the degassed samples. Calculate sample weight as: Degassed sample cell weight–empty sample cell weight.

  • Attach sample cells to analysis ports of the BET apparatus. Verify the level of liquid nitrogen in the Dewar is sufficient for analysis.

  • Using nitrogen as the adsorptive gas analyze the powder samples over the relative pressure range of 0.05–0.30 and use the BET equation to fit the adsorption data.

Results

  • Surface areas of powders produced by spray freezing into liquid ranged from 13 to 134 m2/g.

  • Increasing the feed concentration decreased the specific surface area of the powders.

  • Increasing the feed concentration decreased the submicron particle content.

  • Increasing the droplet size during spray freeze drying resulted in lower specific surface areas.

Method Capsule 6Supersaturation Dissolution Studies Using the Syringe/Filter and Microcentrifuge Methods

Based on the method reported by Curatolo et al. 2009

Objective

  • To determine the ability of HPMCAS in initiation and maintenance of supersaturation of an experimental compound.

Equipment and Reagents

  • Experimental compound CMPD 2

  • HPMCAS-MF

  • 10-mL syringes

  • Model fasted duodenal fluid preheated to 37°C.

  • Oven capable of maintaining 37°C.

  • Wheel capable of rotating syringe in horizontal position at 50 rpm.

  • 20-gauge hypodermic needles

  • 13 mm, 0.45 μm polyvinylidine diflouride syringe filters.

  • Test tubes

  • Polypropylene microcentrifuge tubes

  • Microcentrifuge

  • Vortex mixer

  • Small volume pipette (i.e., 10–100 μL)

  • Diluting solution: 60:40 1.7 wt.% ammonium ascorbate:acetonitrile.

  • HPLC: Phenomenex ultracarb ODS 20 analytical column, PDA detection at 215 nm.

Method

  • Syringe/Filter method:

  • Accurately weigh 7.5 mg of 67% CMPD 2:HPMCAS-MF formulation and add to an empty 10 mL syringe with attached 20-gage needle.

  • Draw 10 mL of model fasted duodenal fluid preheated to 37°C into the syringe via the attached needle.

  • Replace attached needle with 13 mm syringe filter.

  • Shake syringe vigorously for 30 s.

  • Expel six drops of the solution as waste. Collect drops 7–19 as a sample.

  • Draw syringe plunger back to generate an air bubble.

  • Place syringe on rotating wheel (50 rpm) in the 37°C oven.

  • Dilute sample 1:1 with diluting solvent.

  • Repeat sampling procedure at t  =  5, 10, 20, 40, 90, 180 min.

  • Analyze samples on HPLC to quantify CMPD 2.

  • Microcentrifuge method

  • In a 37°C controlled-temperature box, weigh 1.8 mg of formulation into a microcentrifuge tube.

  • Add 1.8 mL of model fasted duodenal fluid to the tube.

  • Close tube, start timer, and vortex mix for 60 s.

  • Transfer tube to microcentrifuge, allow to stand for 6 min, then centrifuge at 13,000  g for 60 s.

  • At the 10 min mark on the timer, remove a 25-μL sample from the supernatant via a pipette. Immediately dilute 1:1 with diluting solution.

  • Resuspend the material via vortex mixing for 30 s.

  • Place tube back in centrifuge. Allow tube to stand undisturbed until the next sampling time point.

  • At each sampling time point, centrifuge the tube for 60 s, remove supernatant sample and resuspend as described. Dilute sample 1:1 with diluting solution.

  • Analyze all samples via HPLC to quantify CMPD 2.

  • Plot the dissolved drug concentration versus time dissolution profile for the API.

Results

  • Compound 2 has an aqueous solubility of 1 μg/mL.

  • The 67% compound 2 solid dispersion formulation with HPMCAS resulted in supersaturation of the test medium.

  • Maximum concentrations of approximately 130 μg/mL were achieved.

  • HPMCAS-MF initiated and maintained supersaturation of compound 2 to a greater extent than HPC, PVAP or the crystalline bulk drug.

  • Both methods proved successful in achieving supersaturation

Method Capsule 7Biphasic Dissolution Testing Utilizing an External Flow through Cell

Based on the method reported by Shi et al. (2010)

Objective

  • To examine the dissolution profiles of three celecoxib formulations using a biphasic dissolution-testing method incorporating an organic phase for drug partitioning.

Equipment and Reagents

  • Octanol

  • Sodium phosphate monobasic monohydrate

  • Sodium hydroxide

  • Gelatin capsules

  • Commercial Celebrex capsules (200 mg dose strength).

  • Extracted celecoxib (extracted from Celebrex capsules via ethanol and subsequent evaporation method).

  • USP Dissolution Apparatus II

  • USP IV flow through cell

  • Piston pump

  • Teflon tubing

  • Modified apparatus II paddle incorporating a second paddle capable of agitating the organic layer during dissolution testing.

  • HPLC (mobile phase 55:45 v/v acetonitrile:ammonium acetate solution).

Method

  • Place 250 mL of 80 mM phosphate buffer (pH 6.8) in the dissolution vessel.

  • Add 200 mL of octanol to the dissolution vessel.

  • Prior to beginning the study, saturate the aqueous phase with octanol and vice versa by agitating the mixture for 30 min.

  • Allow all media to equilibrate to 37°C  ±  0.2°C.

  • Place the Teflon tubing (inlet and outlet) such that the ends are well within the aqueous phase of the dissolution media.

  • Set the paddle speed to 75 rpm.

  • Place the formulation for analysis into the flow through cell.

  • Set the pump flow rate to 30 mL/min.

  • At time points of 15, 30, 45, 60, 75, 90, and 120 min remove a 1-mL sample from the aqueous phase and a 100-μL sample from the organic phase. Do not replace the media.

  • Immediately centrifuge the aqueous phase samples at 14,000 rpm for 6 min. Collect the supernatant for HPLC analysis.

  • Immediately dilute the organic phase samples 100-fold with HPLC mobile phase.

  • Quantitative celecoxib via HPLC analysis; adjustment for dilution mathematically.

Results

  • Corresponding single-phase studies (aqueous; USP Apparatus II) under sink conditions were not discriminatory for formulation performance.

  • Nonsink two-phase dissolution revealed the same formulation performance rank order in the aqueous phase.

  • Analysis of the octanol phase reveals the self-emulsifying drug delivery ­system (SEDDS) outperformed the solution and capsule formulations.

  • SEDDS provided a higher amount of free drug compared to the solution ­formulation in which the drug was associated as surfactant micelles.

  • SEDDS formulation supersaturated the aqueous media under nonsink-biphasic dissolution conditions, allowing greater partitioning into the octanol phase.

  • Aqueous phase AUC values from both single and biphasic dissolution testing exhibited no correlation with in vivo AUC values.

  • Analysis of the octanol phase from biphasic studies revealed a rank-order ­correlation to in vivo results.

Method Capsule 8Supersaturation Dissolution Testing of Amorphous Compositions of a Poorly Water-Soluble Drug

Based on the method reported by DiNunzio et al. 2008

Objective

  • To assess the supersaturation extent and duration of amorphous compositions of itraconazole using concentration enhancing polymers.

Equipment and Reagents

  • Itraconazole; bulk drug and formulations prepared via thin film freezing.

  • Commercial itraconazole capsules (Sporanox).

  • Size 9 porcine gelatin capsules.

  • USP Dissolution Apparatus II with autosampler.

  • 0.1N hydrochloric acid.

  • 0.2M Na3PO4 solution.

  • 0.2 μm PTFE membrane, 13 mm Acrodisc syringe filters.

  • 5 mL syringes

  • HPLC System (5 μm°C18(2) 100  Å, 150 mm  ×  4.6 mm column, flow rate of 1 mL/min, detection at 263 nm).

  • HPLC mobile phase (70:30:0.05 acetonitrile:water:diethanolamine).

  • HPLC vials.

  • Vortex mixer

Method

  • Preheat 735 mL of 0.1N HCl in each dissolution vessel to 37°C.

  • Accurately weigh an amount of formulation equivalent to 37.5 mg of itraconazole. This corresponds to approximately 10Ă— the equilibrium solubility in 0.1N HCl.

  • Pre-wet the weighed powder with 15 mL of heated 0.1N HCl (37°C).

  • Add the prewetted powder slurry to the dissolution vessel.

  • After 2 h, add 250 mL of 0.2 M Na3PO4 to each vessel to bring the pH to 6.8.

  • At time points of 60, 120, 130, 150, 180, 240, 300, 360, and 1,440 min, a 5 mL sample is to be taken via the autosampler without replacement of the withdrawn media.

  • Immediately filter samples following withdraw through a 0.2-μm PTFE membrane.

  • Immediately dilute filtrate 1:1 with mobile phase and vortex mix. Transfer to an HPLC vial.

  • Analyze all samples by HPLC to quantify itraconazole adjusting for the volume change and dilution mathematically.

Results

  • Sporanox pellets were able to rapidly and extensively supersaturate the acidic media. Following the pH transition, the drug rapidly precipitated.

  • Thin film freezing formulations resulted in significantly reduced acidic media concentrations as the polymers used were enteric.

  • Following pH change, cellulose acetate phthalate and polyvinyl acetate phthalate formulations showed the greatest supersaturation in neutral media.

  • Higher ratios of itraconazole:polymer yielded lower degrees of supersaturation.

  • Cellulose acetate phthalate formulations provided longer half-life values for drug in solution, indicating strong concentration enhancing properties.

Method Capsule 9Pulmonary Delivery to the Murine Model Via Dry Powder Insufflation

Based on the method reported by Morello et al. (2009)

Objective

  • To achieve successful pulmonary administration of the tuberculosis vaccine (BCG) to the murine model by dry powder insufflation.

Equipment and Reagents

  • Female BALB/c mice (6–12 weeks old, 18–24 g).

  • BCG vaccine spray-dried powder.

  • Polyethylene tubing (1.19 mm diameter) cut into lengths of approximately 1 cm.

  • Microbalance

  • Penn-Century dry powder insufflator model DP-4 M.

  • Penn-Century model AP-1 air pump.

  • Rodent work stand

  • Lidocaine applicator

  • Mouse-sized speculum

  • Plastic incisor loop

  • Otoscope set

  • Ketamine HCl, xylazine, and acepromazine

  • Yohimbine solution (0.04 mg/mL)

  • Syringe and 21-gauge needle

  • Cage maintained at 37°C.

Method

  • Prepare mixture of ketamine/xylazine/acepromazine by mixing 2 mL ketamine at 100 mg/mL, 0.4 mL of xylazine at 100 mg/mL and 0.6 mL of acepromazine at 10 mg/mL with 7 mL of sterile phosphate–buffered saline.

  • Anesthetize the mouse via an intraperitonial (i.p.) injection (100 μL/20 g body weight) of ketamine/xylazine/acepromazine mixture

  • Load 200–300 μg of powder into the 1 cm segment of polyethylene tubing by dipping the end vertically into the powder bed 4–6 times.

  • Place the filled polyethylene tube into the hole within the insufflator chamber.

  • Attach base of insufflator to the cannula.

  • Place the mouse in the supine position on the mouse work station and place the incisor loop and lateral supports in position.

  • Raise the stand to 60°.

  • Using the otoscope, obtain a clear view of the trachea.

  • Using the speculum to guide the applicator, apply lidocaine to the arytenoid cartilage.

  • Re-obtain visual focus of the trachea with the otoscope.

  • Insert the cannula of the insufflator device into the tracheal opening.

  • Remove the otoscope and attach the air pump.

  • Depress plunger on air pump 4–5 times. Monitor rise and fall the upper chest as confirmation of proper insufflation.

  • Remove mouse from the work stand and give an i.p. injection of 0.11 mg/kg yohimbine solution (0.04 mg/mL)

  • Provide 300 μL 0.9% saline subcutaneously to aid recovery.

  • Move mouse to a 37°C cage until awake.

Results

  • Application of this method allowed rapid administration of the compound relative to similar methods.

  • ≥90% of loaded powder was delivered to the lungs.

  • 91% of the loaded dose reached the lungs.

  • Minor pulmonary damage occurred as a result of the procedure; however, mice were asymptomatic and congestion/hemorrhage resolved over time.

  • The method developed can be modified for use with larger animals by adjusting the dose and air volumes employed.

Method Capsule 10 Oral Drug Delivery of a Poorly Water-Soluble Drug to the Rat Model

Based on the method reported by Wempe et al. 2007

Objective

  • To determine the bioavailability of letrozole complexed with hydroxybutenyl-β-cyclodextrin (HBenβCD) compared to that of the native API via an oral dose administered as a suspension or solution to the rat model.

Equipment and Reagents

  • Letrozole, complexed to HBenβCD and unprocessed.

  • Male and female Sprague–Dawley rats (260–294 and 218–262 g, respectively).

  • 1-mL syringe with 0.01-mL graduations.

  • Sterile glass vials

  • Ethanol

  • Sterile water

  • Sonicator

  • Oral gavage needle (16 gage).

  • Mini-capillary tubes containing EDTA di-potassium salt.

Method

  • Weigh 5.2 mg of letrozole (uncomplexed) into a sterile glass vial.

  • Immediately before dosing, dilute with 12.5% ethanol in water to a concentration of 1 mg/mL using sonication to disperse.

  • Weigh 626.2 mg complexed letrozole-HBenβCD into a glass vial.

  • Immediately before dosing, dilute with 12.5% ethanol in water to generate a 1 mg/mL solution.

  • With the gavage needle attached to the syringe, draw in the desired dose of the suspension or solution to be administered.

  • Holding the rat vertically while supporting the hind legs against the body, insert the gavage needle extending it into the stomach and expel the dose. Immediately remove the gavage needle from the animal.

  • Repeat dosing for all rats.

  • At time points of 0.45, 1.5, 2, 2.6, 2.9, 3.6, 5.1, 6.2, 8.3, 13.9, 24.8, and 36.3 h, collect 125 μL blood samples through the tail vein directly into mini-capillary tubes containing EDTA di-potassium salt.

  • Immediately following blood sample collection, cap the tube and place on dry ice. Keep samples frozen at −80°C until sample preparation for analysis.

Results

  • Letrozole was eliminated from the blood of male rats within 36 h following dosing via oral gavage.

  • The pharmacokinetics of letrozole are strongly gender-dependent.

  • Dosing of letrozole-HBenβCD yielded a twofold increase in the AUC compared to a suspension of the uncomplexed drug.

  • Oral dosing of the complexed formulation increased the Cmax from 87 to 140 ng/mL.

  • The T max was decreased from 8.4 h to 6.3 h by the HBenβCD formulation.

  • Solubility limits the rate and extent of absorption in male rats while only ­limiting the rate of absorption in female rats.

  • Complexation of letrozole with HBenβCD improved oral absorption in male rats while maximizing absorption in female rats

Rights and permissions

Reprints and permissions

Copyright information

© 2012 American Association of Pharmaceutical Scientists

About this chapter

Cite this chapter

O’Donnell, K.P., Williams, R.O. (2012). Optimizing the Formulation of Poorly Water-Soluble Drugs. In: Williams III, R., Watts, A., Miller, D. (eds) Formulating Poorly Water Soluble Drugs. AAPS Advances in the Pharmaceutical Sciences Series, vol 3. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-1144-4_2

Download citation

Publish with us

Policies and ethics