Skip to main content

Injectable Formulations of Poorly Water-Soluble Drugs

  • Chapter
  • First Online:

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

Abstract

A growing number of new therapeutic molecules are limited by low or erratic bioavailability due to poor water solubility. Because of the clinical demand for new and more efficacious anti-cancer, antiviral, and anti-infective drugs, many of these new drugs must be formulated for injection. Poor water solubility can be addressed by a range of formulation approaches such as pH manipulation, salt formation, and cosolvent and surfactant addition, or by more advanced techniques such as complexation, liposomal encapsulation, or nanosuspension. While remaining focused on drug solubility, issues such as buffering, tonicity, sterility, and drug ­product stability also must be considered when formulating injectable products. This chapter outlines a formulator’s approach toward development of an injectable drug product containing an active ingredient with poor solubility. Marketed injectable products, listings of GRAS excipients, and techniques for enhancing solubility are offered as case studies to assist in the formulation process.

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

Buying options

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

Learn about institutional subscriptions

References

  • Akers MJ, Larrimore DS, Guazzo DM (eds) (2003) Parenteral quality control: sterility, pyrogen, particulate, and packaging integrity testing, 3rd edn. Informa Healthcare, New York

    Google Scholar 

  • Akers MJ, Nail SL, Saffell-Clemmer W et al (2007) Top ten hot topics in parenteral science and technology. PDA J Pharm Sci Technol 61(5):337–361

    PubMed  CAS  Google Scholar 

  • Alur H, Jain B, Dey S et al (2005) Physiochemical factors affecting biological activity. In: Ghosh TK, Jasti BR (eds) Theory and practice of contemporary pharmaceutics. CRC Press, Bocan Raton, FL

    Google Scholar 

  • Aschenbrenner DS, Venable SJ (2009) Drugs affecting cardiac rhythm. In: Aschenbrenner DS, Venable SJ (eds) Drug therapy in nursing, 3rd edn. Wolters Kluwer Health, Lippincott Williams & Wilkins, Philadelphia, PA

    Google Scholar 

  • Bari H (2010) A prolonged release parenteral drug delivery system-an overview. Int J Pharm Sci Rev Res 3(1):1–11

    CAS  Google Scholar 

  • Bhalla S (2007) Parenteral delivery. In: Desai A, Lee M (eds) Gibaldi’s drug delivery systems in pharmaceutical care. American Society of Health-Systems Pharmacists, Bethesda,MD

    Google Scholar 

  • Bhattacharjee H, Thoma A (2010) Parenteral drug administration: routes of administration and devices. In: Nema S, Ludwig JD (eds) Pharmaceutical dosage forms parenteral medications, 3rd edn. Informa Healthcare, New York

    Google Scholar 

  • Birrer GA, Merthy SS, Liu J (2001) Parenteral dosage forms. In: Ahuja S, Scypinski S (eds) Handbook of modern pharmaceutical analysis. Academic, San Diego, vol 3

    Google Scholar 

  • Cengage G (2006) Subcutaneous injection. In: Krapp K (ed) Encyclopedia of nursing & allied health. Available via eNotes.com http://enotes.com/nursing-encyclopedia/subcutaneous-injection. Accessed 13 Mar 2011

  • Challa R, Ahuja A, Ali J et al (2005) Cyclodexrins in drug delivery: an updated review. AAPS PharmSciTech 6(2):329–357

    Article  Google Scholar 

  • Croy SR, Kwon GS (2006) Polymeric micelles for drug delivery. Curr Pharm Des 12(36):4669–4684

    Article  PubMed  CAS  Google Scholar 

  • DaunoXome (1996) Product Label. Gilead Sciences San Dimus, CA. http://dailymed.nlm.nih.gov/dailymed/druginfo.cfm?id=628. Accessed 16 Mar 2011

  • De Spiegeleer B, Wattyn E, Slegers G et al (2006) The importance of the cosolvent propylene glycol on the antimicrobial preservative efficacy of a pharmaceutical formulation by DOE-ruggedness testing. Pharm Dev Technol 11(3):275–284

    Article  PubMed  Google Scholar 

  • DeLuca PP, Boylan JC (1992) Formulation of small volume parenterals. In: Avis KE, Lieberman HA, Lachman L (eds) Pharmaceutical dosage forms: parenteral medications, 2nd edn. Mercel Dekker, New York

    Google Scholar 

  • Desai N, Trieu V, Yao Z et al (2006) Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin Cancer Res 12(4):1317–1324

    Article  PubMed  CAS  Google Scholar 

  • Diazemuls (1998) product monograph. In: Gillis MC (ed) CPS Compendium of pharmaceuticals and specialties, 33 rd edn. Canadian Pharmacists Association, Ottawa

    Google Scholar 

  • Ernsberger FM (1959) Attack of glass by chelating agents. J Am Ceram Soc 42(8):373–375

    Article  CAS  Google Scholar 

  • Food and Drug Administration. Safety assessment of Di(2-ethylhexyl)phthalate (DEHP) released from PVC medical devices (2011). Center for devices and radiological health. http://www.fda.gov/downloads/MedicalDevices/ DeviceRegulationandGuidance/GuidanceDocuments/UCM080457.pdf. Accessed 13 Mar 2011

  • Gabizon AA (2001) Stealth liposomes and tumor targeting: one step further in the quest for the magic bullet. Clin Cancer Res 7:223–225

    PubMed  CAS  Google Scholar 

  • Halbert GH (2009) Preformulation. In: Florence AT, Shipman J (eds) Modern phramaceutics: basic principles and systems, vol 1. Informa Healthcare, New York

    Google Scholar 

  • Haldol Injection (2011) Product monograph. Ortho-McNeil-Janssen Pharmaceutcials Raritan, NJ. http://www.drugs.com/pro/haldol-decanoate.html. Accessed 16 Mar 2011

  • Invega Sustenna. Concise monograph (2011). Physician’s desk reference. http://pdr.net/drugpages/concisemonograph.aspx?concise=3070. Accessed 16 Mar 2011

  • Kamat M, DeLuca PP (2010) Formulation development of small and large volume injections. In: Nema S, Ludwig JD (eds) Pharmaceutical dosage forms parenteral medications, 3rd edn. Informa Healthcare, New York

    Google Scholar 

  • Kasra K, Kuzniar AA, Wilson GG et al (1999) Developing an injectable formula containing an oxygen-sensitive drug: a case study ofdanofloxacin injectable. Pharm Dev Technol 4(4):475–480

    Article  Google Scholar 

  • Kerns EH, Di L (2008) Drug-like properties: concepts, structure design and methods: from ADME to tonicity optimization. Academic, London

    Google Scholar 

  • Lambert WJ (2010) Considerations in developing a target product profile for parenteral pharmaceutical products. AAPS PharmSciTech 11(3):1476–1481

    Article  PubMed  CAS  Google Scholar 

  • Lanoxin injection product monograph (2011). RxList. http://www.rxlist.com/lanoxin-drug.htm. Accessed 16 Mar 2011

  • Loftsson T, Jarho P, Másson M et al (2005) Cyclodextrins in drug delivery. Expert Opin Drug Deliv 2(2):335–351

    Article  PubMed  CAS  Google Scholar 

  • McLeish MJ (1992) Comprehensive description. Anal Prof Drug Subst Excip 21:535–572

    Article  Google Scholar 

  • Meyer BK, Ni A, Hu B, Shi L (2007) Antimicrobial preservative use in parenteral products: past and present. J Pharm Sci 96(12):3155–3167

    Article  PubMed  CAS  Google Scholar 

  • Mishra B, Patel BB, Tiwari S (2010) Colloidal nanocarriers: a review on formulation technology types and applications toward targeted drug delivery. Nanomedicine 6:9–24

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  • Shi Y, Porter W, Merdan T, Li LC (2009) Recent advances in intravenous delivery of poorly water-soluble compounds. Expert Opin Drug Deliv 6(12):1261–1282

    Article  PubMed  CAS  Google Scholar 

  • Singh B, Bandopadhyay S, Kapil R et al (2009) Self-emulsifying drug delivery systems (SEDDS): formulation development, characterization, and applications. Crit Rev Ther Drug Carrier Syst 26(5):427–521

    PubMed  CAS  Google Scholar 

  • Sinha VR, Trehan A (2005) Biodegradable microspheres for parenteral delivery. Crit Rev Ther Drug Carrier Syst 22(6):535–602

    PubMed  CAS  Google Scholar 

  • Sinko PJ (2006) Troy DB (eds) Martin’s physical pharmacy and pharmaceutical sciences, 6th edn. Lippincott Williams & Wilkins, Philadelphia

    Google Scholar 

  • Soni MG, Carabin IG, Burdock GA (2005) Safety assessment of esters of p-hydroxybenzoic acid (parabens). Food Chem Tox 43:985–1015

    Article  CAS  Google Scholar 

  • Strickley RG (2004) Solubilizing excipients in oral and injectable formulations. Pharm Res 21(2):201–230

    Article  PubMed  CAS  Google Scholar 

  • Taxol injection (2010) Package insert (2011) Bristol-Myers Squibb Company, Princeton, NJhttp://packageinserts.bms.com/pi/pi_taxol.pdf. Accessed 16 Mar 2011

  • Thompson DO (1997) Cyclodextrins: enabling excipients - their present and future use in pharmaceuticals. Crit Rev Ther Drug Carrier Syst 14(1):1–104

    PubMed  CAS  Google Scholar 

  • Tong W, Wen H (2008) Preformulation aspects of water insoluble compounds. In: Liu R (ed) Water-insoluble drug formulation, 2nd edn. CRC Press, Boca Raton, FL

    Google Scholar 

  • Torchilin VP, Weissig V (eds) (2003) Liposomes: a practical approach, 2nd edn. Oxford, Oxford University Press

    Google Scholar 

  • Tran AX, Whitfield C (2009) Lipopolysaccharides (endotoxins). In: Schaechter M (ed) Encyclodepia of microbiology, 3rd edn. Academic, London, pp 513–528

    Chapter  Google Scholar 

  • Trissel LA (ed) (2002) Handbook of injectable drugs, 12th edn. American Society of Health-System Pharmacists, Bethesda, MD

    Google Scholar 

  • Troy DB (ed) (2006) Remington: the science and practice of pharmacy, 21st edn. Lippincott Williams & Wilkins, Philadelphia

    Google Scholar 

  • USP<788>Particulate matter in injections (2006) The United States Pharmacopeia 29th revision, Rockville

    Google Scholar 

  • van Tellingen O, Huizing MT, Panday VR et al (1999) Cremphor EL causes (pseudo-) non-linear pharmacokinetics of paclitaxel in patients. Br J Cancer 81(2):330–335

    Article  PubMed  Google Scholar 

  • Vemuri N (2010) Preformulation. In: Nema S, Ludwig JD (eds) Pharmaceutical dosage forms parenteral medications, 3rd edn. Informa Healthcare, New York

    Google Scholar 

  • Vivitrol (2010) Full prescribing information. http://vivitrol.com/pdf_docs/prescribing_info.pdf Alkermes Waltham, MA Accessed 16 Mar 2011

  • Wolters Kluwer Health (2006) Top 200 drugs used in hospitals in 2006. Pharmaceutical Audit Suite (PHAST)

    Google Scholar 

  • Wong J, Brugger A, Khare A et al (2008) Suspensions for intravenous (IV) injection: a review of development, preclinical and clinical aspects. Adv Drug Deliv Rev 60(8):939–954

    Article  PubMed  CAS  Google Scholar 

Capsule References

  • Dong Y, Ng WK, Surana U et al (2008) Solubilization and preformulation of poorly water soluble and hydrolysis susceptible N-epoxymethyl-1,8-naphthalimide (ENA) compound. Int J Pharm 356(1):130–136

    Article  PubMed  CAS  Google Scholar 

  • Kelmann RG, Kuminek G, Teixeira HF et al (2007) Carbamazepine parenteral nanoemulsions prepared by spontaneous process. Int J Pharm 342:231–239

    Article  PubMed  CAS  Google Scholar 

  • Levy MY, Benita S (1989) Design and characterization of a submicronized o/w emulsion of diazepam for parenteral use. Int J Pharm 54:103–112

    Article  CAS  Google Scholar 

  • Li P, Tabibi E, Yalkowsky SH (1999a) Solubilization of flavopiridol by pH control combined with cosolvents, surfactants, or complexants. J Pharm Sci 88(9):945–947

    Article  PubMed  CAS  Google Scholar 

  • Li P, Zhao L, Yalkowsky SH (1999b) Combined effect of cosolvent and cyclodextrin on solubilization of nonpolar drugs. J Pharm Sci 88(11):1107–1111

    Article  PubMed  CAS  Google Scholar 

  • Xiong R, Lu W, Li J et al (2007) Preparation and characterization of intravenously injectable nimodipine nanosuspension. Int J Pharm 350(1–2):338–343

    PubMed  Google Scholar 

  • Zhong H, Deng Y, Wang X et al (2005) Multivesicular liposome formulation for the sustained delivery of breviscapine. Int J Pharm 301:15–24

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michael P. Boquet .

Editor information

Editors and Affiliations

Appendices

Method Capsule 1Solubilization of a Poorly Water-Soluble Drug Using a Surfactant with pH Adjustment

Based on the method reported by Li et al. (1999a).

Objective

  • To increase the solubility of flavopiridol, an antineoplastic agent with a water solubility of 0.025 mg/mL, by changing the solution pH and incorporating a surfactant.

Equipment and Reagents

  • Flavopiridol (apparent pK a 5.86).

  • Polysorbate 80 (PS80).

  • Citrate buffer.

  • End-over-end mechanical rotator operating at 20 rpm.

  • Pinnacle octylamine HPLC column (150 cm x 4.6 mm).

  • Mobile phase of 0.1% triethylamine in 50 mM phosphate buffer pH 2.5 and acetonitrile in a ration of 35:65 (flow rate 1 mL/min; detection 263 nm).

  • Syringe filter unit (0.45 μm).

  • pH meter.

Method

  • Prepare solutions of varying concentrations of PS80 (0, 2.5, 5, 10, and 20%) in citrate buffer with a pH of 4.3 and 8.4.

  • Perform solubility study by placing 0.5 mL of each solution into vials with excess flavopiridol.

  • Attach vials to rotator, place at 25°C, and rotate at 20 rpm for 6 days or until equilibrium solubility is achieved. (Note: The drug is stable for >2 months under these conditions).

  • Filter samples through 0.45-μm syringe filter membrane and check final solution pH.

  • Perform HPLC analysis of samples for potency (solubility) of flavopiridol.

Results

  • The solubility of flavopiridol in solution without surfactant was 1.37 mM and 0.055 mM at pH 4.3 and 8.4, respectively.

  • The solubility of a drug in the micelle is influenced by the micellar partition coefficient and drug water solubility.

  • Increasing the concentration of PS80 resulted in a substantial linear increase in drug solubility. The increase was considerably greater (up to ∼27 mM at 20% PS80) at pH 4.3, where the majority of drug is ionized.

  • Solubilization of the ionized drug by PS80 is more critical than that of the unionized species for increasing the overall solubility due to the increased water solubility resulting from the increased solubility of drug in the micelles.

Method Capsule 2Solubilization of a PWS Drug using a Combination of Cosolvent and Surfactant

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

Objective

  • To develop a stable injectable solution of N-epoxymethyl-1,8-naphthalimide, a nonionizable PWS drug (0.0116 mg/mL) that is very susceptible to hydrolytic degradation by screening a number of solubilization techniques.

Equipment and Reagents

  • N-epoxymethyl-1,8-naphthalimide (ENA).

  • Cosolvents: ethanol, polyethylene glycol 400 (PEG-400), PG, and glycerol.

  • Surfactants: Cremophor EL and Tween 80.

  • Complexation agents: HPβCD and hydroxypropyl-γ-cyclodextrin (HPγCD).

Method

  • Determine the solubility of ENA in cosolvent–water solutions with concentrations up to 50% (w/v).

  • Determine the solubility of ENA in surfactant–water solutions with concentrations up to 20% (w/v).

  • Determine the solubility of ENA in aqueous solutions containing up to 20% (w/v) cyclodextrins.

  • Based on the results from the above solubility studies, determine the solubility of ENA in nonaqueous solution containing different ratios of the best performing solubilizers: Cremophor EL and ethanol (0:100, 50:50, 60:40, 70:30, and 100:0% v/v).

  • Select a number of ratios of Cremophor EL:ethanol from the previous stability study and assess formulation stability upon dilution with saline.

  • Assess formulation physical and chemical stability at different storage conditions.

Results

  • Aqueous solutions containing concentration of 50% ethanol, PEG400, PG, or glycerol have an ENA solubility of 0.531, 0.269, 0.181, and 0.038 mg/mL, respectively.

  • The solubility of ENA is increased to 0.395 and 0.410 mg/mL with 20% of Tween 80 and Cremophor EL, respectively.

  • The solubility of ENA is increased to 0.169 and 0.190 mg/mL with 20% of HPβCD and HPγCD, respectively.

  • Cremophor EL:ethanol ratios of 0:100, 50:50, 60:40, 70:30, and 100:0 increased the solubility of ENA to 1.25, 3.90, 4.54, 5.42, and 6.65, respectively.

  • Of all the ratios screened, only a combination of 70% Cremophor EL and 30% ethanol containing 4 mg/mL of ENA did not precipitate upon dilution (5- to 20-fold) with saline. This formulation is physically and chemically stable for over 4 months at either 5°C or room temperature.

Method Capsule 3Solubilization Using a Combination of Complexation Agent and Cosolvent

Based on the method reported by Li et al. (1999b).

Objective

  • To increase the solubility of fluasterone, a nonpolar oncology compound, using various amounts of cosolvent (ethanol) in combination with the complexation agent hydroxyl propyl-β-cyclodextrin (HPβCD).

Equipment and Reagents

  • Fluasterone (intrinsic solubility 0.045 μg/mL)

  • Ethanol

  • HPβCD

  • End-over-end mechanical rotator operating at 20 rpm

  • Pinnacle octylamine HPLC column (150 cm x 4.6 mm)

  • Mobile phase of 75% acetonitrile in water (flow rate 1.1 mL/min; detection 220 nm)

  • Syringe filter unit (0.45 μm)

  • pH meter

Method

  • Prepare solutions of varying concentrations of ethanol (0–70%) and HPβCD (0–20%).

  • Place 0.5 mL of each solution into glass vials with excess flavopiridol.

  • Connect vials to rotator, place at 25°C, and rotate at 20 rpm for 6 days or until equilibrium solubility is achieved. (Note: The drug is stable for >50 days under these conditions).

  • Filter samples through a 0.45-μm syringe filter membrane and check final solution pH.

  • Perform HPLC analysis of samples for potency of fluasterone.

Results

  • An exponential increase in the solubility of fluasterone is observed in a cosolvent and water system with increasing ethanol concentration.

  • Using HPβCD in an aqueous solution yields a linear increase in drug solubility corresponding with increasing HPβCD concentration.

  • As ethanol concentration increases from 0.2 to 25.06% in the presence of HPβCD, the polarity of the solvent decreases. The result is reduced drug complexation within the HPβCD cavity and a decrease in the drug solubility over this range, with a minimum at 25.06%.

  • At high ethanol concentrations (25.06–75%), the solubility increases due to the increase in the concentration of free drug and ternary complex.

  • With the ethanol concentration held constant (>25.06%), there is a linear increase in drug solubility with increasing amounts of HPβCD.

Method Capsule 4Preparation of a Submicronized Injectable Emulsion Formulation

Based on the method reported by Levy and Benita (1989).

Objective

  • To design and characterize an emulsion formulation for diazepam, which meets the requirements for an IV or IM administration.

Equipment and Reagents

  • Diazepam

  • Purified soybean oil

  • Egg yolk phospholipids (purified fractionated complex emulsifier)

  • Poloxamer (nonionic emulsifier)

  • Glycerin (osmotic agent)

  • Methyl and butyl p-hydroxybenzoic acid (preservative)

  • α-Tocopherol (antioxidant)

  • Sodium hydroxide solution (10%)

  • High-shear mixer (Polytron)

  • Two-stage pressure homogenizer (Gaulin homogenizer)

Method

  • The formulation is composed of 0.5% (w/w) diazepam, 20% oil, 1.2% phospholipids, 2% poloxamer, 2.25% glycerin, 0.02% α-tocopherol, 0.2% methyl and 0.075% butyl p-hydroxybenzoic acid, and water for injection to 100 g.

  • Perform preparation and processing with nitrogen atmosphere and sterile conditions.

  • Add poloxamer, glycerin, and preservatives to the aqueous phase and dissolve.

  • Dissolve phospholipids and diazepam in the oil phase containing α-tocopherol.

  • Filter both phases, and heat separately to 70°C while mixing with a magnetic stirrer. Increase the temperature to 85°C for the emulsification step using the high-shear mixer.

  • Once the emulsion is formed, cool rapidly. A reduced droplet size is achieved using the two-stage Gaulin homogenizer. Adjust pH to the desired level with sodium hydroxide and filter the emulsion.

Results

  • Characterization of the formulation including the oil phase properties, pH, phase/volume ratio, emulsifier, phospholipid, nonionic emulsifier, and diazepam concentrations produced an optimized stable emulsion.

  • Droplet size was reduced to an acceptable size for IV administration from a mean size of 0.65 μm by a high-shear mixer to 0.27 μm, using the Gaulin two-stage pressure homogenizer.

Method Capsule 5Preparation of Multivesicular Liposome Formulation for Sustained IM Delivery

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

Objective

  • To design a multivesicular liposome (MVL) sustained delivery formulation for IM injection.

Equipment and Reagents

  • Breviscapine (bioactive ingredient)

  • Phosphatidycholine

  • Phosphatidylglycerol

  • Triolein or tricaprylin

  • Cholesterol

  • Sucrose and glucose

  • Buffers: 50 mM arginine buffer pH 7, 40 mM L-Lysine

  • Chloroform–diethyl ether (1:1 v/v)

  • T 18 basic Ultra-Turrax mixer

Method

  • Use the double-emulsion process to produce breviscapine MVLs. Prepare a lipid mixture in 1 mL chloroform–diethyl ether with 40 mg phosphatidycholine, 8 mg phosphatidylglycerol, 40 mg cholesterol, and triolein or ­tricaprylin at a 5.75:1 molar ratio of phosphatidycholine to total triglyceride content.

  • Prepare an aqueous solution containing 40 mg/mL of breviscapine, 4% w/v sucrose in 50 mM arginine buffer pH 7.

  • Emulsify the lipid and aqueous solutions to make a W/O emulsion at 10,000 rpm for 8 min with the mixer.

  • Prepare the second aqueous solution with 40 mM L-Lysine and 3.4% glucose and emulsify with the W/O emulsion to form a W/O/W emulsion. Transfer to an Erlenmeyer flask and remove the chloroform–diethyl ether by nitrogen flushing the liquid surface at 30°C.

  • Centrifuge at 600g for 5 min to remove free breviscapine from the MVLs and resuspend in a buffered saline solution.

Results

  • Brevicapine MVLs produced by this method significantly prolonged the release of drug both in vitro (5–6 days) and in vivo (IM injection in rats lasted 4–5 days) compared to other the liposome preparation techniques investigated.

Method Capsule 6Preparation of Nanoemulsions for IV Delivery

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

Objective

  • To prepare a nanoemulsion for IV delivery of a PWS drug, carbamazepine.

Equipment and Reagents

  • Carbamazepine (CBZ)

  • Ethanol

  • Acetone

  • Soybean lecithin (lipophilic emulsifier)

  • Medium chain triglycerides (MCT)

  • Polyoxyl 35 castor oil (lipophilic emulsifier)

  • Polysorbate 80 (hydrophilic emulsifier)

  • Glycerol (tonicity agent)

  • Magnetic stirrer

Method

  • Mix CBZ (final emulsion concentration of 2 mg/mL) with castor oil or a 1:1 mixture of castor oil:MCT (w/w) and prepare the lipophilic emulsifier ­(soybean lecithin or polyoxyl 35 castor oil at 6% w/w) by dissolving it in a 50:50 mixture of acetone:ethanol (v/v).

  • Make the oil phase by adding the prepared emulsifier to the drug:oil dispersion.

  • Prepare the aqueous phase by dissolving 4% (w/w) PS80 and 2.5% (w/v) glycerol in water.

  • While stirring (moderate speed) with a magnetic stirrer, slowly add the oil phase to the aqueous phase to form the nanoemulsion.

  • Remove solvent and water by reducing the pressure, adjust pH to 7.0 with 0.1 M sodium hydroxide, and store the emulsion at 5°C.

Results

  • The emulsion droplets are spherically shaped, have an amorphous core, and range in size from 100 to 250 nm.

  • The resulting optimized nanoemulsion formulation demonstrates 3 months ­stability based on droplet size, polydispersity, zeta potential, and drug content data.

  • The drug nanoemulsion meets requirements for IV administration and will be tested for in vivo evaluation.

Method Capsule 7Preparation of an IV Nanosuspension Formulation for Reduced Irritation and Phlebitis upon Injection

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

Objective

  • To improve upon a current clinical IV formulation for nimodipine, which contains a high concentration of ethanol that causes injection site pain, irritation, and phlebitis.

Equipment and Reagents

  • Nimodipine (compound for the treatment of subarachnoid hemorrhage-related vasospasm)

  • Sodium deoxycholate

  • Poloxamer 188

  • Mannitol

  • Polysorbate 80

  • MC One (fluid jet mill)

  • Niro-Soavi NS1001L (high-pressure homogenizer)

  • Laboratory freeze drier

Method

  • Perform all the following steps under reduced lighting conditions to protect the light-sensitive drug.

  • Jet mill the coarse powder of nimodipine to produce a microparticulate powder.

  • Disperse with a magnetic stirrer 0.5% (w/v) the milled powder in an aqueous solution composed of 0.6% (w/v) poloxamer 188, 0.4% (w/v) sodium cholic acid, and 4.0% (w/v) mannitol.

  • Perform pre-milling using the high-pressure homogenizer (maintain sample temperature at 25–30°C) by starting with the settings at 200 bar with two cycles then increasing to 500 bar with five cycles.

  • Follow the pre-milling step with 15–20 cycles at 1,500 bar to produce the nanosuspension.

  • Lyophilize the nanosuspension by drying for 15 hr at −15°C (below 200 mTorr), with a secondary step of 3 hr at −5°C, and a final step of 2 hr at 20°C.

  • Sterilize by gamma irradiation for 6 hr with an absorbed dose of 12 kGy.

Results

  • Injection toleration studies performed in rabbits demonstrate that this formulation reduces occurrence of phlebitis and minimizes local irritation when compared with the clinical ethanol-based product.

Rights and permissions

Reprints and permissions

Copyright information

© 2012 American Association of Pharmaceutical Scientists

About this chapter

Cite this chapter

Boquet, M.P., Wagner, D.R. (2012). Injectable Formulations 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_6

Download citation

Publish with us

Policies and ethics