Skip to main content

Solid-State Techniques for Improving Solubility

  • Chapter
  • First Online:
Formulating Poorly Water Soluble Drugs

Part of the book series: AAPS Advances in the Pharmaceutical Sciences Series ((AAPS,volume 3))

Abstract

Poor aqueous solubility of a drug substance can often be attributed to strong intermolecular forces within its crystal lattice which, in turn, prevent molecules from escaping in solution. Through the use of solid-state chemistry, it is possible to modify the crystal structure in such a way that mitigates intermolecular forces, thus improving aqueous solubility and increasing rates of dissolution. Solid-state techniques utilized for solubility enhancement include the formation of salts, polymorphic or amorphous forms, and co-crystals. Each technique has specific advantages and, in some cases, disadvantages that may prevent its successful use. The purpose of this chapter is to describe each of the methods, allowing the reader to gain an understanding of solid-state modifications available for solubility enhancement.

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

  • Aguiar AJ, Zelmer JE (1969) Dissolution behavior of polymorphs of chloramphenicol palmitate and mefenamic acid. J Pharm Sci 58:983–987

    Article  PubMed  CAS  Google Scholar 

  • Aguiar AJ, Krc J, Kinkel AW, Samyn JC (1967) Effect of polymorphism on the absorption of chloramphenicol from chloramphenicol palmitate. J Pharm Sci 56:847–853

    Article  PubMed  CAS  Google Scholar 

  • Allesø M, van den Berg F, Cornett C, Jørgensen FS, Halling Sørensen B, de Diego HL, Hovgaard L, Aaltonen J, Rantanen J (2008) Solvent diversity in polymorph screening. J Pharm Sci 97:2145–2159

    Article  PubMed  Google Scholar 

  • Alvarez AJ, Singh A, Myerson AS (2009) Polymorph screening: comparing a semi-automated approach with a high throughput method. Cryst Growth Des 9:4181–4188

    Article  CAS  Google Scholar 

  • Bastin RJ, Bowker MJ, Slater BJ (2000) Salt selection and optimisation procedures for pharmaceutical new chemical entities. Org Process Res Dev 4:427–435

    Article  CAS  Google Scholar 

  • Bauer J, Spanton S, Henry R, Quick J, Dziki W, Porter W, Morris J (2001) Ritonavir: an extraordinary example of conformational polymorphism. Pharm Res 18:859–866

    Article  PubMed  CAS  Google Scholar 

  • Bechtloff B, Nordhoff S, Ulrich J (2001) Pseudopolymorphs in industrial use. Cryst Res Tech 36:1315–1328

    Article  CAS  Google Scholar 

  • Berge SM, Bighley LD, Monkhouse DC (1977) Pharmaceutical salts. J Pharm Sci 66:1–19

    Article  PubMed  CAS  Google Scholar 

  • Bis JA, Vishweshwar P, Weyna D, Zaworotko MJ (2007) Hierarchy of supramolecular synthons: ersistent hydroxyl pyridine hydrogen bonds in cocrystals that contain a cyano acceptor. Mol Pharm 4:401–416

    Article  PubMed  CAS  Google Scholar 

  • Black SN, Collier EA, Davey RJ, Roberts RJ (2007) Structure, solubility, screening, and synthesis of molecular salts. J Pharm Sci 96:1053–1068

    Article  PubMed  CAS  Google Scholar 

  • Blagden N, Md M, Gavan PT, York P (2007) Crystal engineering of active pharmaceutical ingredients to improve solubility and dissolution rates. Adv Drug Deliv Rev 59:617–630

    Article  PubMed  CAS  Google Scholar 

  • Bowker MJ, Stahl PH (2008) Preparation of water soluble compounds through salt formation. In: Wermuth CG (ed) The practice of medicinal chemistry. Academic, New York, p 749

    Google Scholar 

  • Byrn SR, Pfeiffer RR, Stowell JG (1999a) Drugs as molecular solids. Solid-state chemistry of drugs. SSCI, West Lafayette, pp 143–241

    Google Scholar 

  • Byrn SR, Pfeiffer RR, Stowell JG (1999b) Polymorphs. Solid-state chemistry of drugs. SSCI, West Lafayette, pp 143–241

    Google Scholar 

  • Chemburkar SR, Bauer J, Deming K, Spiwek H, Patel K, Morris J, Henry R, Spanton S, Dziki W, Porter W, Quick J, Bauer P, Donaubauer J, Narayanan BA, Soldani M, Riley D, McFarland K (2000) Dealing with the impact of ritonavir polymorphs on the late stages of bulk drug process development. Org Process Res Dev 4:413–417

    Article  CAS  Google Scholar 

  • Corrigan OI (2006) Salt forms: pharmaceutical aspects. In: Swarbrick J (ed) Encyclopedia of pharmaceutical technology. Informa Healthcare, New York, pp 3177–3187

    Google Scholar 

  • Dannenfelser R-M, He H, Joshi Y, Bateman S, Serajuddin ATM (2004) Development of clinical dosage forms for a poorly water soluble drug I: application of polyethylene glycol–polysorbate 80 solid dispersion carrier system. J Pharm Sci 93:1165–1175

    Article  PubMed  CAS  Google Scholar 

  • Elder DP, Delaney E, Teasdale A, Eyley S, Reif VD, Jacq K, Facchine KL, Oestrich RS, Sandra P, David F (2010) The utility of sulfonate salts in drug development. J Pharm Sci 99:2948–2961

    PubMed  CAS  Google Scholar 

  • Engel GL, Farid NA, Faul MM, Richardson LA, Winneroski LL (2000) Salt form selection and characterization of LY333531 mesylate monohydrate. Int J Pharm 198:239–247

    Article  PubMed  CAS  Google Scholar 

  • Etter MC, Adsmond DA (1990) The use of cocrystallization as a method of studying hydrogen bond preferences of 2-aminopyrimidine. Journal of the Chemical Society, Chemical Communications, pp 589–591

    Google Scholar 

  • Etter MC, Reutzel SM, Choo CG (1993) Self-organization of adenine and thymine in the solid state. J Am Chem Soc 115:4411–4412

    Article  CAS  Google Scholar 

  • Fleischman SG, Kuduva SS, McMahon JA, Moulton B, Bailey Walsh RD, Rodríguez-Hornedo N, Zaworotko MJ (2003) Crystal engineering of the composition of pharmaceutical phases: ­multiple-component crystalline solids involving carbamazepine. Cryst Growth Des 3:909–919

    Article  CAS  Google Scholar 

  • Gardner CR, Almarsson O, Chen H, Morissette S, Peterson M, Zhang Z, Wang S, Lemmo A, Gonzalez-Zugasti J, Monagle J, Marchionna J, Ellis S, McNulty C, Johnson A, Levinson D, Cima M (2004) Application of high throughput technologies to drug substance and drug product development. Comput Chem Eng 28:943–953

    Article  CAS  Google Scholar 

  • Good DJ, Rodríguez-Hornedo Nr (2009) Solubility advantage of pharmaceutical cocrystals. Cryst Growth Des 9:2252–2264

    Article  CAS  Google Scholar 

  • Gould PL (1986) Salt selection for basic drugs. Int J Pharm 33:201–217

    Article  CAS  Google Scholar 

  • Grant DJW, Higuchi T (1990) Solubility, intermolecular forces, and thermodynamics. In: Saunders WH (ed) Solubility behavior of organic compounds, vol XXI. Wiley-Interscience, New York, pp 12–88

    Google Scholar 

  • Gross TD, Schaab K, Ouellette M, Zook S, Reddy JP, Shurtleff A, Sacaan AI, Alebic-Kolbah T, Bozigian H (2007) An approach to early-phase salt selection: Application to NBI-75043. Org Process Res Dev 11:365–377

    Article  CAS  Google Scholar 

  • Gupta P, Chawla G, Bansal AK (2004) Physical stability and solubility advantage from amorphous celecoxib: the role of thermodynamic quantities and molecular mobility. Mol Pharm 1:406–413

    Article  PubMed  CAS  Google Scholar 

  • Haleblian J, McCrone W (1969) Pharmaceutical applications of polymorphism. J Pharm Sci 58:911–929

    Article  PubMed  CAS  Google Scholar 

  • Hancock BC (2002) Disordered drug delivery: destiny, dynamics and the Deborah number. J Pharm Pharmacol 54:737–746

    Article  PubMed  CAS  Google Scholar 

  • Hancock BC, Parks M (2000) What is the true solubility advantage of the different forms? Pharm Res 17:397–404

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  • Hancock BC, Shamblin SL, Zografi G (1995) Molecular mobility of amorphous pharmaceutical solids below their glass transition temperatures. Pharm Res 12:799–806

    Article  PubMed  CAS  Google Scholar 

  • Hickey MB, Peterson ML, Scoppettuolo LA, Morrisette SL, Vetter A, Guzmán H, Remenar JF, Zhang Z, Tawa MD, Haley S, Zaworotko MJ, Almarsson Ö (2007) Performance comparison of a co-crystal of carbamazepine with marketed product. Eur J Pharm Biopharm 67:112–119

    Article  PubMed  CAS  Google Scholar 

  • Huang L-F, Tong W-Q (2004) Impact of solid state properties on developability assessment of drug candidates. Adv Drug Deliv Rev 56:321–334

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  • Jain N, Yang G, Machatha SG, Yalkowsky SH (2006) Estimation of the aqueous solubility of weak electrolytes. Int J Pharm 319:169–171

    Article  PubMed  CAS  Google Scholar 

  • Kim J-S, Kim M-S, Park HJ, Jin S-J, Lee S, Hwang S-J (2008) Physicochemical properties and oral bioavailability of amorphous atorvastatin hemi-calcium using spray-drying and SAS process. Int J Pharm 359:211–219

    Article  PubMed  CAS  Google Scholar 

  • Kobayashi Y, Ito S, Itai S, Yamamoto K (2000) Physicochemical properties and bioavailability of carbamazepine polymorphs and dihydrate. Int J Pharm 193:137–146

    Article  PubMed  CAS  Google Scholar 

  • Kumar L, Amin A, Bansal AK (2007) An overview of automated systems relevant in pharmaceutical salt screening. Drug Discov Today 12:1046–1053

    Article  PubMed  CAS  Google Scholar 

  • Kuroda R, Imai Y, Tajima N (2002) Generation of a co-crystal phase with novel coloristic properties via solid state grinding procedures. Chem Commun 2848–2849

    Google Scholar 

  • Lang M, Kampf JW, Matzger AJ (2002) Form IV of carbamazepine. J Pharm Sci 91:1186–1190

    Article  PubMed  CAS  Google Scholar 

  • Lee S, Hoff C (2002) Large-scale aspects of salt formation: processing of intermediates and final products. In: Stahl PH, Wermuth CG (eds) Pharmaceutical salts: properties, selection, and use. Wiley-VCH, New York, pp 191–220

    Google Scholar 

  • Leuner C, Dressman J (2000) Improving drug solubility for oral delivery using solid dispersions. Eur J Pharm Biopharm 50:47–60

    Article  PubMed  CAS  Google Scholar 

  • Li S, Wong S, Sethia S, Almoazen H, Joshi YM, Serajuddin ATM (2005) Investigation of solubility and dissolution of a free base and two different salt forms as a function of pH. Pharm Res 22:628–635

    Article  PubMed  CAS  Google Scholar 

  • Lipinski CA (2002) Poor aqueous solubility: industry wide problem in drug discovery. Am Pharmaceut Rev 5:82–85

    Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  • Loftsson T, Brewster ME (1996) Pharmaceutical applications of cyclodextrins. 1. Drug solubilization and stabilization. J Pharm Sci 85:1017–1025

    Article  PubMed  CAS  Google Scholar 

  • Mao C, Pinal R, Morris KR (2005) A quantitative model to evaluate solubility relationship of polymorphs from their thermal properties. Pharm Res 22:1149–1157

    Article  PubMed  CAS  Google Scholar 

  • McNamara D, Childs S, Giordano J, Iarriccio A, Cassidy J, Shet M, Mannion R, O’Donnell E, Park A (2006) Use of a glutaric acid cocrystal to improve oral bioavailability of a low solubility API. Pharm Res 23:1888–1897

    Article  PubMed  CAS  Google Scholar 

  • Medina C, Daurio D, Nagapudi K, Alvarez-Nunez F (2010) Manufacture of pharmaceutical co-crystals using twin screw extrusion: a solvent-less and scalable process. J Pharm Sci 99:1693–1696

    PubMed  CAS  Google Scholar 

  • Miller DA, DiNunzio JC, Yang W, McGinity JW, Williams RO III (2008) Targeted intestinal delivery of supersaturated itraconazole for improved oral absorption. Pharm Res 25:1450–1459

    Article  PubMed  CAS  Google Scholar 

  • Morissette SL, Soukasene S, Levinson D, Cima MJ, Almarsson Ö (2003) Elucidation of crystal form diversity of the HIV protease inhibitor ritonavir by high-throughput crystallization. Proc Natl Acad Sci USA 100:2180

    Article  PubMed  CAS  Google Scholar 

  • Morissette SL, Almarsson Ö, Peterson ML, Remenar JF, Read MJ, Lemmo AV, Ellis S, Cima MJ, Gardner CR (2004) High-throughput crystallization: polymorphs, salts, co-crystals and solvates of pharmaceutical solids. Adv Drug Deliv Rev 56:275–300

    Article  PubMed  CAS  Google Scholar 

  • Morris KR, Fakes MG, Thakur AB, Newman AW, Singh AK, Venit JJ, Spagnuolo CJ, Serajuddin A (1994) An integrated approach to the selection of optimal salt form for a new drug candidate. Int J Pharm 105:209–217

    Article  CAS  Google Scholar 

  • O’Connor KM, Corrigan OI (2001) Preparation and characterisation of a range of diclofenac salts. Int J Pharm 226:163–179

    Article  PubMed  Google Scholar 

  • Parks GS, Huffman HM, Cattoir FR (1928) Studies on glass II: the transition between the glassy and liquid states in the case of glucose. J Phys Chem 32:1366–1379

    Article  CAS  Google Scholar 

  • Parks GS, Snyder LJ, Cattoir FR (1934) Studies on glass XI. Some thermodynamic relations of glassy and alpha-crystalline glucose. J Phys Chem 2:595–598

    Article  CAS  Google Scholar 

  • Paulekuhn GS, Dressman JB, Saal C (2007) Trends in active pharmaceutical ingredient salt selection based on analysis of the orange book database. J Med Chem 50:6665–6672

    Article  PubMed  CAS  Google Scholar 

  • Peterson ML, Morissette SL, McNulty C, Goldsweig A, Shaw P, LeQuesne M, Monagle J, Encina N, Marchionna J, Johnson A, Gonzalez-Zugasti J, Lemmo AV, Ellis SJ, Cima MJ, Almarsson Ö (2002) Iterative high-throughput polymorphism studies on acetaminophen and an experimentally derived structure for form III. J Am Chem Soc 124:10958–10959

    Article  PubMed  CAS  Google Scholar 

  • Porter WW III, Elie SC, Matzger AJ (2008) Polymorphism in carbamazepine cocrystals. Crys Growth Des 8:14–16

    Article  CAS  Google Scholar 

  • Pudipeddi M, Serajuddin ATM (2005) Trends in solubility of polymorphs. J Pharm Sci 94:929–939

    Article  PubMed  CAS  Google Scholar 

  • Remenar JF, Morissette SL, Peterson ML, Moulton B, MacPhee JM, Guzmán HR, Almarsson Ö (2003) Crystal engineering of novel cocrystals of a triazole drug with 1,4-dicarboxylic acids. J Am Chem Soc 125:8456–8457

    Article  PubMed  CAS  Google Scholar 

  • Rodríguez-Spong B, Price CP, Jayasankar A, Matzger AJ, Rodríguez-Hornedo N (2004) General principles of pharmaceutical solid polymorphism: a supramolecular perspective. Adv Drug Deliv Rev 56:241–274

    Article  PubMed  Google Scholar 

  • Schultheiss N, Newman A (2009) Pharmaceutical cocrystals and their physicochemical properties. Crys Growth Des 9:2950–2967

    Article  CAS  Google Scholar 

  • Serajuddin ATM (2007) Salt formation to improve drug solubility. Adv Drug Deliv Rev 59:603–616

    Article  PubMed  CAS  Google Scholar 

  • Shalaev E, Zografi G (2002) The concept of “structure” in amorphous solids from the perspective of the pharmaceutical sciences. Amorphous Food and Pharmaceutical Systems 281:11–30

    Article  CAS  Google Scholar 

  • Shan N, Zaworotko MJ (2008) The role of cocrystals in pharmaceutical science. Drug Discov Today 13:440–446

    Article  PubMed  CAS  Google Scholar 

  • Shattock TR, Arora KK, Vishweshwar P, Zaworotko MJ (2008) Hierarchy of supramolecular synthons: persistent carboxylic acid-pyridine hydrogen bonds in cocrystals that also contain a hydroxyl moiety. Cryst Growth Des 8:4533–4545

    Article  CAS  Google Scholar 

  • Singhal D, Curatolo W (2004) Drug polymorphism and dosage form design- a practical perspective. Adv Drug Deliv Rev 56:335–347

    Article  PubMed  CAS  Google Scholar 

  • Tong WQT, Whitesell G (1998) In situ salt screening-a useful technique for discovery support and preformulation studies. Pharm Dev Technol 3:215–223

    Article  PubMed  CAS  Google Scholar 

  • Torchilin V (2007) Micellar nanocarriers: pharmaceutical perspectives. Pharm Res 24:1–16

    Article  PubMed  CAS  Google Scholar 

  • Trask AV, Motherwell WDS, Jones W (2004) Solvent-drop grinding: green polymorph control of cocrystallisation. Chem Commun 7:890–891

    Article  Google Scholar 

  • Trask AV, Motherwell WDS, Jones W (2005a) Pharmaceutical cocrystallization: engineering a remedy for caffeine hydration. Cryst Growth Des 5:1013–1021

    Article  CAS  Google Scholar 

  • Trask AV, van de Streek J, Motherwell WDS, Jones W (2005b) Achieving polymorphic and stoichiometric diversity in cocrystal formation: importance of solid-state grinding, powder x-ray structure determination, and seeding. Cryst Growth Des 5:2233–2241

    Article  CAS  Google Scholar 

  • Ware E, Lu DR (2004) An automated approach to salt selection for new unique trazodone salts. Pharm Res 21:177–184

    Article  PubMed  CAS  Google Scholar 

  • Wells JI (1988) Pharmaceutical preformulation: the physicochemical properties of drug substances. Ellis Horwood, Chichester

    Google Scholar 

  • Xiang T-X, Anderson BD (2004) A molecular dynamics simulation of reactant mobility in an amorphous formulation of a peptide in poly(vinylpyrrolidone). J Pharm Sci 93:855–876

    Article  PubMed  CAS  Google Scholar 

  • Yu L (1995) Inferring thermodynamic stability relationship of polymorphs from melting data. J Pharm Sci 84:966–974

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  • Yu L, Reutzel SM, Stephenson GA (1998) Physical characterization of polymorphic drugs: an integrated characterization strategy. Pharmaceut Sci Tech Today 1:118–127

    Article  CAS  Google Scholar 

  • Zhang GGZ, Henry RF, Borchardt TB, Lou X (2007) Efficient co-crystal screening using solution-mediated phase transformation. J Pharm Sci 96:990–995

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Justin R. Hughey .

Editor information

Editors and Affiliations

Appendices

Method Capsule 1Preparation of Amorphous Solids - Spray-Drying

Based on the method reported by Kim et al. (2008).

  • Objective

  • To obtain an amorphous drug substance by the spray-drying technique

Equipment and Reagents

  • Atorvastatin calcium

  • Acetone or tetrahydrofuran

  • Laboratory-scale spray-dryer

Method

  • Dissolve atorvastatin calcium in acetone or tetrahydrofuran at a concentration of 100 mg/mL.

  • Set the drying air flow rate to 0.70 m3/min.

  • Set the inlet temperature of the spray dryer to 70°C.

  • Feed the solution into the spray dryer at 3 mL/min with an atomization pressure of 10 kPa.

  • Ensure that the outlet temperature is in the range of 62–65°C.

  • Collect the dried powder.

Results

  • Laser diffraction analysis revealed that the particle size of atorvastatin calcium spray-dried from acetone and tetrahydrofuran was 3.62  ±  0.15 μm and 7.31  ±  0.21 μm, respectively.

  • BET specific surface area analysis demonstrated that the surface area of atorvastatin particles spray-dried from acetone and tetrahydrofuran was 3.69  ±  0.06 m2/g and 0.95  ±  0.03 m2/g, respectively. The specific surface area of unprocessed material was 14.56  ±  0.17 m2/g.

  • Differential scanning calorimetry and X-ray diffraction analyzes indicated that all spray-dried material lacked crystalline character and was amorphous.

Method Capsule 2Preparation of Amorphous Solids - Supercritical Anti-solvent Processing

Based on the method reported by Kim et al. (2008).

Objective

  • To obtain an amorphous drug substance by supercritical anti-solvent (SAS) processing.

Equipment and Reagents

  • Atorvastatin calcium

  • Acetone or tetrahydrofuran

  • Laboratory-scale supercritical anti-solvent processor

  • Carbon dioxide (CO2)

Method

  • Deliver CO2 into particle formation vessel equilibrated at 40°C until the pressure reaches 12 MPa.

  • Dissolve atorvastatin calcium in acetone or tetrahydrofuran at a concentration of 100 mg/mL.

  • The drug solution and supercritical CO2 were co-injected through a two-flow nozzle at 0.5 g/min and 45 g/min, respectively.

  • After the drug solution was exhausted, fresh CO2 was cycled into the vessel to remove residual solvent at 45 g/min.

Results

  • Laser diffraction analysis revealed that the particle size of SAS-processed atorvastatin particles from acetone and tetrahydrofuran was 68.7  ±  15.8 nm and 95.7  ±  12.2 nm, respectively. Particle size of the unprocessed material was 3.83  ±  0.08 μm.

  • BET-specific surface area analysis demonstrated that the surface area of SAS processed atorvastatin particles from acetone and tetrahydrofuran was 120.35  ±  1.40 m2/g and 79.78  ±  0.93 m2/g, respectively. The specific surface area of unprocessed material was 14.56  ±  0.17 m2/g.

  • Differential scanning calorimetry and X-ray diffraction results indicated that all spray-dried material lacked crystalline character and was amorphous.

Method Capsule 3Preparation of Amorphous Solids - Melt Quenching

Based on the method reported by Hancock and Parks (2000).

Objective

  • To obtain an amorphous drug substance by melt quenching.

Equipment and Reagents

  • Indomethacin

  • Liquid nitrogen

Method

  • Heat indomethacin to a temperature that induces melting (∼160°C).

  • Quench the melt with liquid nitrogen such that indomethacin solidifies.

Results

  • Differential scanning calorimetry and X-ray diffraction results indicated that melt-quenched material lacked crystalline character and was amorphous.

Method Capsule 4Preparation of Co-crystals - Temperature-Induced Precipitation

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

Objective

  • To obtain a co-crystal of carbamazepine and saccharin by temperature-induced precipitation.

Equipment and Reagents

  • Carbamazepine

  • Saccharin

  • Ethanol

  • Methanol

  • Water-jacketed glass crystallization vessel

Method

  • Anhydrous carbamazepine (0.089 mol) and saccharin (0.089 mol) were ­combined in the crystallization vessel.

  • Solids were dissolved in 280 mL of a 62.5/37.5% (v/v) ethanol/methanol, mixture and heated to 70°C for 1 h under reflux.

  • Temperature was decreased in 10°C increments while stirring to induce precipitation.

  • Following equilibration at 30°C, solids were isolated using a Buchner funnel and rinsed with cold ethanol.

  • The resulting powder was air-dried.

Results

  • A product yield of 76% was obtained.

  • Microscopy studies showed that the particle size of the crystals was between 500 and 1,000 μm.

  • X-ray powder diffraction and differential scanning calorimetry studies demonstrated that a single polymorphic co-crystal form was prepared.

Method Capsule 5Preparation of Co-crystals - Seed-Induced Precipitation

Based on the method reported by McNamara et al. (2006).

Objective

  • To obtain co-crystals of a new drug candidate (compound 1) with glutaric acid by seed-induced precipitation.

Equipment and Reagents

  • Compound 1

  • Glutaric acid

  • Chloroform

  • Water-jacketed glass crystallization vessel

Method

  • Compound 1 (8.431 mmol) and glutaric acid (8.410 mmol) were dissolved in boiling chloroform with stirring.

  • The solution was concentrated by continued boiling until the volume was 50 mL.

  • Co-crystal seeds (generated in thermal experiments) were introduced into the hot solution.

  • After crystallization began, the solution was cooled over a 15 min period.

  • Approximately 100 mL of cyclohexane was added and the solution was cooled on ice for 30 min.

  • The co-crystal was isolated by filtration.

  • The resulting powder was air-dried.

Results

  • A product yield of 92% was obtained.

  • The volumetric median diameter Dv(0.5) and that of the 90th percentile Dv(90) were found to be 49 μm and 131 μm, respectively.

  • X-ray diffraction analysis showed a unique pattern that was distinguished from compound 1 and glutaric acid.

  • Differential scanning calorimetry experiments indicated that the co-crystal exhibited a melting point different from that of compound 1 or glutaric acid.

Method Capsule 6 Preparation of Co-crystals - Grinding

Based on the method reported by Trask et al. (2004)

Objective

  • To obtain co-crystals of caffeine and glutaric acid by a solid-state grinding technique.

Equipment and Reagents

  • Anhydrous caffeine

  • Glutaric acid

  • Optional non-polar solvents: n-hexane, cyclohexane, or heptane

  • Optional polar solvents: chloroform, dichloromethane, acetonitrile and water

  • Ball grinder/mill

Method

  • Equimolar amounts of caffeine and glutaric acid were combined in a stainless steel grinding jar.

  • Optional: Add four drops of either a non-polar solvent or a polar solvent.

  • Grind the materials together.

  • Allow any residual solvent to evaporate.

Results

  • X-ray diffraction analysis demonstrated that co-crystals were formed when the material was prepared in the absence of solvent and with non-polar or polar solvents.

  • Form I of the co-crystal was found to form when no solvent or a non-polar solvent was used.

  • Form II was predominantly formed when a polar solvent was used.

Rights and permissions

Reprints and permissions

Copyright information

© 2012 American Association of Pharmaceutical Scientists

About this chapter

Cite this chapter

Hughey, J.R., Williams, R.O. (2012). Solid-State Techniques for Improving Solubility. 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_3

Download citation

Publish with us

Policies and ethics