Dipen Patel*, ST Prajapati, CN Patel, Jatin Patel and Vachhani Savan
Shri Sarvajanik Pharmacy College, Near Arvind Baug, Mehsana-384 001, India
*Corresponding Author E-mail: dipeneforever@yahoo.co.in
ABSTRACT:
Insoluble Drug Delivery technology, which has been successfully, addressed the problem of water-insoluble drug delivery. Water-insoluble drugs pose intricate problems in their formulation and delivery. Poorly water-soluble drugs traditionally have been formulated for oral administration through their micronization. Micronization increases their in vivo dissolution rates by reducing particle size and increasing surface area with a concomitant gain in bioavailability. New approaches in formulating poorly soluble drugs, such as the use of surface stabilized nano or microparticles, inclusion in polymer or lipophilic matrices such as nanospheres, hydrophobic carrier systems, self-dispersible systems, and molecular complexation with agents suitable for lipophilic drugs, have demonstrated significant improvements. Hydrophobic carrier systems or self-dispersible systems can be employed only for those drugs that are sufficiently soluble in the carrier. Similarly, a matrix-inclusion system can be employed if the extent of drug loading and the drug release profile within the gastrointestinal tract are acceptable. Insoluble Drug Delivery technology formulations have displayed high drug payload, low amount of free drug in the continuous phase, almost all of the drug present in the dispersed particulate phase, no chemical change in the drug caused by the formulation process, absence of drug-vehicle interaction, narrow particle size distribution with well-defined particle morphology, a variety of suspensions and solid dosage forms, excellent bioavailability when required, and long formulations shelf-lives.
KEYWORDS: IDD technology, Advantage, Future Direction.
INTRODUCTION:
Drug insolubility is one of the most intractable problems in new drug development and in reformulation of existing medications. Insoluble Drug Delivery (IDD) technology, which has been successfully, addressed the problem of water-insoluble drug delivery. Example formulations employing IDD technology are discussed, particularly with respect to their ability to improve oral bioavailability.
Water-insoluble drugs pose intricate problems in their formulation and delivery. Water insolubility of many drugs is often manifested in poor gastrointestinal absorption and bioavailability, intra- and interindividual bioavailability variations, and food interaction in their absorption after oral administration1-3. Some of the emerging oral drug delivery systems that have addressed the drug insolubility problems are summarized in Table 1.
Poorly water-soluble drugs traditionally have been formulated for oral administration through their micronization by air-jet milling4. Micronization in creases their in vivo dissolution rates by reducing particle size and increasing surface area with a concomitant gain in bioavailability4,5.
New approaches in formulating poorly soluble drugs, such as the use of surface stabilized nano or microparticles6–15, inclusion in polymer or lipophilic matrices such as nanospheres16–21, hydrophobic carrier systems22–24, self-dispersible systems 25–29, and molecular complexation with agents suitable for lipophilic drugs 30,31, have demonstrated significant improvements. Each technique, although displaying specific advantages, has characteristic limitations narrowing its suitability to only certain types of drugs. For instance, hydrophobic carrier systems or self-dispersible systems can be employed only for those drugs that are sufficiently soluble in the carrier. Similarly, a matrix-inclusion system can be employed if the extent of drug loading and the drug release profile within the gastrointestinal tract are acceptable.
While molecular complexes with hydrophilic carriers for water-insoluble drug delivery are well developed, e.g., cyclodextrin complexes30, water dispersible molecular aggregates, such as micelles and liposomes, provide interesting, albeit largely unexplored, alternatives. Transport of liposomes or other colloidal particulate systems via paracellular and transcellular routes from normal epithelial tissue or Peyer’s patches leads to different outcomes of drug delivery and immunization, respectively24,32. Formulating water-insoluble drugs with polymerized, microencapsulated, polymer-coated, or targeted liposomes, together with a greater understanding of their cellular processing, will ultimately lead to effective therapies from oral liposomes24.
Lipophilic drugs are harbored into liposomal bilayers with very low drug: vehicle ratios10,11, thus limiting their utility only to high-potency insoluble drugs of vaccines.
A phospholipid-based drug delivery system for water-insoluble drugs, termed IDD10,11 (formerly known as MicroDroplet6,7 and MicroCrystal8 technologies) is believed to be the first example to enable administration of highly concentrated drug substances as surface-modified microparticle formulations. Similar delivery systems exploiting these principles have been described subsequently9,12–15. Various IDD sub technologies are described in Table 2. Previously reviewed IDD formulations for parenteral administration10,11 include both IDD-PTM, i.e., insoluble drug delivery of microparticle formulations, and IDD-DTM i.e., insoluble drug delivery as an aqueous dispersion of submicrometer-size droplets.
IDD technologies can address insoluble drug delivery problems across various dosage forms, including oral, topical and pulmonary. The principal inert ingredients used in IDD formulations are biocompatible and have been previously accepted by the US FDA. IDD formulations can offer the following benefits33.
a. Lower toxicity formulations.
b. Sustained-release depot formulations.
c. Improved oral bioavailability.
d. Oral, intravenous or ophthalmic formulations of drugs that currently are available only in other dosage forms.
e. High drug payloads.
IDD®-P (MicroParticle)
A microparticulate variation of the IDD® drug delivery system, which consists of a pure solid drug in the core of the particle.
IDD®-D formulations (MicroDroplet)
The core is constituted of essentially liquid drug substance.
IDD®-P and IDD®-D formulations are produced by application of high shear, cavitation or impaction (e.g., attrition, homogenisation, microfluidisation, milling, ultrasonication, etc.) to reduce the drug particle size in the presence of phospholipids (and/or other surface modifiers) that associate at the freshly generated drug surface. A particle size reduction from approximately 100–200 µm to about 1 µm is achieved resulting in a very homogeneous and stable formulation.
IDD®-SE (Self-Emulsifying)
The IDD®-SE technology constitutes a special class of the IDD® systems due to their production. The particles of IDD®-P and IDD®-D formulations result from application of a physical or mechanical process. On the other hand, surface stabilised micrometre to submicrometre sized particles or droplets are self generated when a dosage form containing IDD®-SE formulation is exposed to an aqueous medium such as those present in gastro-intestinal or vascular compartments.
IDD-P FORMULATIONS:
The IDD approach to insoluble drug delivery involves formulation of microparticles of water-insoluble drugs stabilized by surface-modifying agents such as phospholipids with or without other surface modifiers. Size reduction of drug particles dispersed in aqueous medium in the presence of phospholipids (and/or other surface modifiers) results in association of the latter with the newly cleaved plane of the freshly generated drug surface. In Figure 134, the electron micrographs display drug raw material as about 100 µm crystals, and an IDD-P formulation as about 0.5 µm surface-modified microparticles.
Structure:
The use of phospholipids in the production of IDD formulations as multiphasic aqueous dispersions calls for a comparison between the IDD technology and liposomes. The distinctive feature is the presence of a hydrophilic core and phospholipid bilayers in liposomes versus a solid or liquid water-insoluble drug core and several phospholipid microstructural domains in the IDD particles (Figure 2)10. The IDD system consists of a submicrometer-sized water-insoluble drug core stabilized with phospholipids with or without other surface modifiers. IDD-P formulations, a microparticulate variation of the IDD drug delivery system, consist of pure solid drug in the core of the particle. Similarly, in the IDD-D formulations (the microdroplet variation) the core is constituted of essentially liquid drug substance. Three distinct phospholipid domains are thought to exist in these formulations. The phospholipid molecules of Domain A are considered closely associated with the drug core. This domain may consist of the phospholipid molecular coating on the drug core. Domain B may be composed of more phospholipid layers and/or small unilamellar vesicles loosely associated with the core. Multiple phospholipid bilayers, typical of multilamellar liposomes, seem to be absent in the IDD-P or IDD-D formulations as evidenced by their X-ray diffraction and DSC profiles (unpublished results). Within the suspensions Domain B is thought to move with the drug core and gives the particle its hydrodynamic size. Domain C is composed of small bilayer vesicles and/ or other phospholipid microstructures freely dispersed in the aqueous vehicle. A small fraction of the formulated drug may remain partitioned in the phospholipid microstructures of Domains B and C. In the case of IDD-D formulations, depending on the phospholipid solubility in the liquid drug, a small and undetermined amount of phospholipid may be dissolved in the lipophilic liquid drug core. On the other hand, liposomes are phospholipid bilayer vesicles. Lipophilic drugs are usually formulated with multilamellar vesicle liposomes composed of many concentric bilayers (only two bilayers are shown here). The lipophilic drug molecule is incorporated in the phospholipid bilayers that are separated by aqueous medium.
Table 1: Some Emerging Oral Drug Delivery Systems for Water-Insoluble or Poorly Water-Soluble Drugs
Formulation technology |
Characteristics |
Micronization Traditional micronization[4,5] |
Particle size reduction, typically with an air-jet mill. Dissolution rate enhancement via increased surface area. High drug payload. Bioavailability improvement.
|
Surface-stabilized small particle technology Insoluble Drug Delivery (IDD) technology[6–11] |
Surface stabilized submicrometer size particles with biocompatible and safe phospholipids and other surface modifiers. Enhancement of dissolution rate via increased surface area. High drug payload. Bioavailability improvement. Uses GRAS-listed traditional excipients.
|
NanoCrystal technology[12–14]
|
Surface stabilized particles of ca. 400 nm. Dissolution rate enhancement via increased surface area. High drug payload. Bioavailability improvement.
|
Nanosuspensions[15]
|
Surface stabilized particles of submicrometer size. Dissolution rate enhancement via increased surface area and increased saturation solubility. Bioavailabilityimprovement.
|
Matrix inclusion technology Nanospheres and microspheres[16–18] |
Submicrometer to micrometer size particles of biodegradable polymeric matrix with entrapped drug. Drug loading and release depend on solubility of the drug in polymeric matrix material. |
Nanosol technology[19] |
Collagen- or gelatin-based drug matrices, 150–250-nm particles containing insoluble drugs. |
Egalet CR system[20]
|
Dispersion of drug in water-degradable polymer matrix. Capable of sustained delivery via oral as well as long term delivery into urinary bladder or vagina.
|
INDAS: insoluble drug absorption system[21] |
Stabilized amorphous form of drug displays improved dissolution rate and absorption.
|
Hydrophobic carrier systems Solid lipid nanoparticles (SLN)[22], and emulsions[23] |
High-pressure emulsification of drug dissolved in melted lipid followed by cooling to form solid particles with drug in the core or associated on the particle surface. Special case of oil-in-water (O/W) emulsion where the oil carrier is solid at ambient temperature. Restricted by low drug loading. O/W emulsions of liquid drug of lipophilic drug dissolved in oil are not very pragmatic for oral delivery because large-volume administration is required.
|
Liposomes[24] |
Lipophilic drugs can be incorporated in the bilayers of liposomes. Very low drug loading. Limited mainly to parenteral and topical administration. Possible vehicle for oral vaccines.
|
Self-dispersible systems Self emulsifying or microemulsifying drug delivery systems[25–27] |
Active ingredient(s) dissolved in a carrier system of a lipophilic phase, surfactant and cosurfactant, and a hydrophilic phase. On mixing with gastric fluid makes O/W type emulsion or microemulsion giving rise to higher bioavailability. Restricted by the solubility of the drug in the vehicle and dispersibility in gastric fluid. |
Table 2 IDD Sub-technologies and Their Characteristics
IDD technologya |
Characteristics |
Administration route and examples |
IDD-PTM
|
Submicrometer-size particulate formulation stabilized with phospholipid with or without other sur face modifiers. Steam-sterilized suspensions for parenteral administration. Can be g-irradiated to sterilize in dry state. Applicable to broad range of drugs. |
Oral (CTM650, cyclosporine) Parenteral (itraconazole, bupivacaine, and antineoplastic drugs: busulfan and 9-nitrocamptothecin) Inhalation (budesonide)
|
IDD-DTM
|
Steam sterilized oil-in-water emulsion. Essentially pure water-insoluble liquid drug in the dispersed oil phase. Takes advantage of oil solubility of some water-insoluble solid drugs.
|
Parenteral (propofol,cyclosporine)
|
IDD-SETM and IDD-METM
|
Self-emulsifiable or self micro-emulsifiable solution in a carrier containing lipophilic solvent and surfactant.Forms submicrometer-size emulsion or microemulsion on exposure to gastrointestinal environment. Takes advantage of drug solubility in oil and surfactant mixture.
|
Oral (CTM650, cyclosporine)
|
IDD-ITM
|
Incorporates IDD-P particulate compositions within a biodegradable implantable carrier. Long-acting preparation of water-insoluble drugs. Applicable to a broad range of drugs.
|
Implantable (flurbiprofen, bupivacaine) Topical |
IDD-OTM
|
Micrometer to submicrometer-size particulate suspension of oil-insoluble drugs in a pharmaceutically active oily medium packaged within capsules or gelcaps. Site-specific delivery through enteric coating. Applicable to drugs that are insoluble in water and oil. |
Oral (budesonide)
|
Where “a” These technologies are currently being applied to formulate a number of water-insoluble new chemical entities and to reformulate existing medicines.
Drug-Vehicle Interaction:
The association of phospholipid molecules with the drug surface originates primarily from weak interactions such as hydrophobic, van der Waals, dipolar, or combinations thereof. Minimal (if any) chemical interaction exists between the phospholipid sheath and the drug core. A phospholipid palisade structure has been illustrated by determinations of phase transition temperatures typical of the phospholipid structures in IDD formulations35. Absence of strong interaction between the phospholipids and the drug entities in these formulations was also illustrated by 19F-NMR chemical shift experiments36.
MANUFACTURING PROCESS:
Fundamental unit operations that can be employed to generate small particles of IDD-P systems include (a) particle fracture processes and (b) particle nucleation processes. High shear, cavitation, or impaction (e.g., milling, attrition, homogenization, microfluidization, etc.) is employed for the drug particle fracture. Alternatively, ultrasmall drug nuclei are generated from a pressurized fluid solution of the drug substance in the second process. The presence of the phospholipid and/ or other surface modifiers is essential for the formation and stability of the IDD-P systems by both processes. Association of phospholipid with or without other surface modifiers onto the freshly generated drug surface provides a capsule domain that prevents particle growth by aggregation, flocculation, agglomeration, or Ostwald ripening during production and shelf life.
Figure 1 IDD micronization.
The IDD micronization process produces homogeneous suspensions of surface-stabilized submicrometer-sized particles of water-insoluble drugs. Electron micrograph A shows 50–100 µm particles of a native model drug with a water solubility of approximately 40 µg/ml (distance between the markers ~51.7 µm). The IDD process yields surface-stablized submicrometer-sized drug particles, as shown in electron micrograph B (distance between markers ~447 nm). Surface stabilization may be elicited by phospholipids alone or mixtures of phospholipids and other surfactants.
Particle Size Reduction:
Acceptability of microfluidization as a unit operation in the pharmaceutical industry has been very well established. For instance, many large-scale pharmaceutical processes based on microfluidizers are reported for liposomes, emulsions, nanospheres, and cell-rupture-related unit operations37–41. Many processes in the chemical industry have been reported that use a microfluidizer for solid particle size reduction41. While merits of this unit process for surface-modified particulate suspensions have been discussed43, particle size reduction of water insoluble drugs by microfluidization appears to be a pioneering adaptation of this process at pilot as well as commercial scale manufacture of IDD-P formulations.
Figure 2: Schematic illustration of the IDD system and lipsome.
Domain A are considered closely associated with the drug core. Domain B may be composed of more phospholipid layers and/or small unilamellar vesicles loosely associated with the core. Domain C is composed of small bilayer vesicles and/ or other phospholipid microstructures freely dispersed in the aqueous vehicle.
Particle Growth from Solution:
Several elegant, very simple, yet easily scalable supercritical fluid (SCF)-based processes have been developed for IDD-P formulations that employ controlled growth of solid drug nuclei from solution phase rather than size reduction unit operations. For example, stable microparticle suspensions of IDD-P cyclosporine (CyA), a water-insoluble drug, have been produced by rapid expansion of SCF solution to aqueous solution or suspension (RESAS)44,45.
In an alternative approach, the product of a rapid expansion of liquefied gas solution (RELGS) of water-insoluble drugs can be optionally homogenized (RELGS-H process) at high pressure46. The homogenization step is thought to enhance the surface modifier interaction with the surface of the IDD-P particles and possibly further generate the necesary microstructures of the phospholipids typical of IDD formulations.
Solid Dosage Forms:
Suitability of the drying unit operations for IDD-P suspensions has been further established by reconstitution of dried formulations with aqueous media10,47. For instance, IDD-P suspensions of cyclosporine, and itraconazole prepared by microfluidization were lyophilized and reconstituted with simulated gastric fluid to original particle size distribution. Agents such as sucrose, α.α-trehalose, and mannitol were added to protect the microstructure of these formulations during lyophilization.
In certain cases where liquid suspensions of IDD-P formulations may display instability over extended periods of time, a lyophilized or otherwise dried product provides preferred dosage forms that allow the maintenance of particle size upon reconstitution. For example, lyophilized IDD-P bupivacaine11 and busulfan48 formulations have been developed that can be easily reconstituted with Water-for-Injection prior to administration.
PHARMACEUTICAL CHARACTERISTICS:
Stability:
Depending on the drug, IDD formulations display excellent physical and chemical stability. As a example, particle size of an IDD-P itraconazole formulation suffered negligible change under a variety of stress conditions, including freeze/thaw, thermal cycling between 2–8°C and 40°C, shaking, sedimentation by centrifugation, lyophilization followed by reconstitution, and even steam sterilization10.
Release Profile and Bioavailability:
The combination of the physical characteristics of each of the components of an IDD-P formulation offers a unique opportunity to control the pharmacokinetics and drug release mechanism of the formulated suspension. Thus, IDD-P formulations can display rapid release profiles when formulated with appropriate excipients and provide examples of improvement in bioavailability of water-insoluble drugs.
SAFETY AND REGULATORY ISSUES:
They have a long history of use and safety in pharmaceutical formulations. Large oral dosages of phospholipids of up to 80g/day have been reported49. As a result of their endogenous nature in mammalian systems, phospholipids are acceptable to regulatory agencies for oral as well as parenteral administration. Formulations containing phospholipids have toxicities comparable to or less than those of unformulated drug and are highly tissue compatible.
COMPETITIVE ADVANTAGE AND FUTURE DIRECTIONS50:
The reliance on a major physical property of the drug, i.e., its water insolubility, rather than on a specific chemical property, facilitates application of IDD technology to a broad range of insoluble drugs. Advantage can be taken of the spectrum of preparation scales available in IDD technology for the formulation and evaluation of a wide variety of both new and old compounds. Classes of water-insoluble compounds include newly discovered and promising water-insoluble drugs, proprietary drugs with excellent potential that remain shelved in pharmaceutical libraries because of perceived or prior inhouse difficulties with their formulation, and currently marketed drugs that may be otherwise less than adequately formulated for successful delivery or for which revised and improved formulation by IDD technology may offer improved patent life. Once a viable drug formulation is achieved on a small scale, it can be scaled up with little difficulty. Small-scale preparation capabilities in IDD technology make it a useful tool for relatively rapid in vivo evaluation of water-insoluble new chemical entities that result from in vitro research and often remain unexplored owing to lack of viable formulation. IDD technology encompasses insoluble drugs that are not limited by therapeutic and chemical classes, does not exclude lipid-soluble or lipid-insoluble drugs, and provides safe and efficacious products for a variety of administration routes. In addition to the inherent safety profiles associated with phospholipid particle stabilizers, IDD technology formulations have displayed high drug payload, low amount of free drug in the continuous phase, almost all of the drug present in the dispersed particulate phase, no chemical change in the drug caused by the formulation process, absence of drug-vehicle interaction, narrow particle size distribution with well-defined particle morphology, a variety of suspensions and solid dosage forms, excellent bioavailability when required, and long formulations shelf-lives.
Phase I and Phase II clinical trials have been completed with various IDD formulations for oral as well as injectable10,11 administration. These have demonstrated excellent safety and efficacy profiles. Beyond bioavailability improvement of water-insoluble drugs by virtue of the small size of their particles and depending on the drug, IDD-P formulations used for oral administration can be formulated to modulate gastrointestinal absorption in part by the intervention of the biocompatible formulation components.
Excellent reproducibility in scale up for manufacturing IDD-formulated products has been achieved. Unlike liposomes, these formulations have not encountered any major scale up problems. Formulating with traditional and GRAS listed phospholipid excipients and the absence of any vehicle for formulation related side effects is expected to facilitate regulatory approval of IDD formulations.
CONCLUSION:
Insoluble Drug Delivery technology, which has been successfully, addressed the problem of water-insoluble drug delivery and development of IDD formulation useful to improved bioavailability, bioequivalence at lower dose levels, and for increased efficiency in drug delivery.
ACKNOWLEDGEMENT:
My deep heartfelt thanks to Dr. D. M. Patel, Principal, Parul Institute of Pharmacy, Baroda, for the valuable guidance for the completion of present Review work.
REFERENCES:
1. Fleisher D, Li C, Zhou Y, Pao LH, Karim A. Drug, meal and formulation interactions influencing drug absorption after oral administration: clinical implications. Clin Pharmacokinet 1999: 36:233–254.
2. Ponto LL, Schoenwald RD. Furosemide (frusemide): A pharmacokinetic /pharmacodynamic review (Part I). Clin Pharmacokinet 1990:18:381–408.
3. Posti J, Katila K, Kostiainen T. Dissolution rate limited bioavailability of flutamide, and in vitro–in vivo correlation. Eur J Pharm Biopharm 2000:49:35–39.
4. Abdou HM. Methods for enhancement of drug dissolution characteristics. Dissolution, Bioavailability & Bioequivalence, Easton, Mack Publishing Company, 1989, 265–284.
5. Ansel HC, Popovich NG. Dosage from design. In, Ansel HC, Popovich NG(ed). Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia, Lea & Febiger 1990: 56–58
6. Haynes DH. Microdroplets of water insoluble drugs. International patent W085/ 00011 1985.
7. Haynes DH. Method of inducing local anesthesia using microdroplets of a general anesthetic. US patent 4,622,219, 1986.
8. Haynes DH. Phospholipid-coated microcrystals: injectable formulations of waterinsoluble drugs. US patents 5,091,187 and 5,091,188, 1992.
9. Haynes DH. Microdroplets of water-insoluble drugs and injectable formulations containing same. US patent 4,725,442, 1988.
10. Pace SN, Pace GW, Parikh I, Mishra AK. Novel injectable formulations of insoluble drugs. Pharm Technol 1999:23:116–134.
11. Mishra A, Robinson G, Pace G. Novel injectable formulations of water-insoluble drugs. Controlled Release 2000:17:21–30.
12. Liversidge GG, Cundy KC, Bishop JF, Czekai DA. Surface modified drug nanoparticles. US patent 5,145,684, 1992.
13. Illig KJ, Sarpotdar P. Formulations comprising Olin 10-G to prevent particle aggregation and increase stability. US patent 5,340,564, 1994.
14. Bosch HW, Marcera DM, Mueller RL, Swanson JR, Mishra DS. Process for preparing therapeutic compositions containing nanoparticles. US patent 5510118, 1996.
15. Muller RH, Becker R, Kruss B, Peters K. Pharmaceutical nanosuspensions for medicament administration as systems with increased saturation solubility and rate of solution. US patent 5,858,410, 1999.
16. Speiser PP. Nanoparticles and liposomes: a state of the art. Methods Find Exp Clin Pharmacol 1991:13:337-342.
17. Kreuter J. Nanoparticles. In, Kreuter J (ed). Colloidal Drug Delivery Systems, New York, Marcel Dekker, 1994;219–342.
18. Okada H, Toguchi H. Biodegradable microspheres in drug delivery. Crit Rev Ther Drug Carr Syst 1995;12:1–99.
19. Wunderlich JC, Schick U, Werry J, Freidenreich J. Gelatin or collagen hydrolysate containing drug formulation that provides for immediate release of nanoparticle drug compounds. US patent 5,932,245, 1999.
20. Bar-Shalom D, Kindt-Larsen T. Controlled release erodible composition. US patent 5,618,560, 1997. See also http://www.egalet.dk/home.htm.
21. Devane JG, Mulligan S, Kavanagh M, Davis S et al. New developments in sustained-release antihypertensive therapy, formulation and pharmacokinetic considerations. Am J Cardiol 1992;69:23E–27E. See also
http:// www.elan.ie/drugdel/technologies/oral/indas.asp.
22. Muller RH, Mader K, Gohla S. Solid lipid nanoparticles (SLN) for controlled drug delivery, A review of the state of the art. Eur J Pharm Biopharm 2000;50(1):161–177.
23. Santos MNS, Benita S, Seiller M. Oral emulsions: preparation and bioavailability. J Pharm Belg 1993;48:211–226.
24. Rogers JA, Anderson KE. The potential of liposomes in oral drug delivery. Crit Rev Ther Drug Carr Syst 1998;15:421–480.
25. Gershanik T, Benita S. Self-dispersing lipid formulations for improving oral absorption of lipophilic drugs. Eur J Pharm Biopharm 2000;50:179–188.
26. Tenjarla S. Microemulsions: an overview and pharmaceutical applications. Crit Rev Ther Drug Carr Syst 1999;16:461–521.
27. Serajuddin AT. Solid Dispersion of poorly water-soluble drugs: Early promises, subsequent problems and recent breakthoughs. J Pharm Sci 199988:1058–1066..
28. Klyashchitsky BA, Owen AJ. Drug delivery systems for cyclosporine: achievements and complications. J Drug Target 1998;5:443–458.
29. Constantinides PP. Lipid microemulsions for improving drug dissolution and oral absorption: physical and biopharmaceutical aspects. Pharm Res 1995;12:1561–1572.
30. Thompson DO. Cyclodextrins-enabling excipients: their present and future use in pharmaceuticals. Crit Rev Ther Drug Carr Syst 1997;14:1–104.
31. Aungst BJ. Novel formulation strategies for improving oral bioavailability of drugs with poor membrane permeation or presystemic metabolism. J Pharm Sci 1993;82:979– 987.
32. Genta I, Perugini P, Pavanetto F, Modena T et al. Microparticulate drug delivery systems 1999;87:305–313.
34. Rathbone MJ et l. Modified-Release Drug Delivery Technology, New York, Marcel Dekker, 2003;157.
35. Lansman J, Haynes DH. Kinetics of a CA2_-triggered membrane aggregation reaction of phospholipid membranes. Biochim Biophys Acta 1975;394:335–347.
36. Haynes DH, Kirkpatrick AF. Ultra-long duration local anesthesia produced by intradermal injection of lecithin-coated methoxyflurane microdroplets. Proc Int Symp Control Rel Bioact Mater 1987;14:293–294.
37. Vivier A, Vuillemard JC, Ackermann HW, Poncelet D. Large-scale blood substitute production using a microfluidizer. Biomater Artif Cells Immobiliz Biotechnol 1992;20: 377-397.
38. White MD, Marcus D. Disintegration of microorganisms. Adv Biotechnol Processes 1988;8:51–96.
39. Lidgate DM, Trattner T, Shultz RM, Maskiewicz R. Sterile filtration of a parenteral emulsion. Pharm Res 1992;9:860–863.
40. Bodmeier R, Chen H, Tyle P, Jarosz P. Spontaneous formation of drug-containing acrylic nanoparticles. J Microencapsul 1991;8:161–170.
41. Lidgate DM, Fu RC, Byars NE, Foster LC, Fleitman JS. Formulation of vaccine adjuvant muramyldipeptides, Processing optimization, characterization, and bioactivity of an emulsion vehicle. Pharm Res 1989;6:748–752.
42. http://www.microfluidicscorp.com/mi_ref.htm.
43. Illig KJ, Mueller RL, Ostrander KD, Swanson JR. Use of microfluidizer processing for preparation of pharmaceutical suspensions. Pharm Technol 1996;20:78–88.
44. Henriksen IB, Mishra AK, Pace GW, Johnston KP et al. Insoluble drug delivery. WIPO WO9714407A1, 1997.
45. Young TJ, Mawson S, Johnstron KP, Henriksen IB et al. Rapid expansion from supercritical to aqueous solution to produce submicron suspension of water-insoluble drugs. Biotechnol Progr 2000;16:402–407.
46. Pace GW, Vachon MG, Mishra AK, Henriksen IB et al. Processes to generate submicron particles of water-insoluble compounds. WIPO WO9965469A2, 1999.
47. Parikh I, Mishra AK, Donga R. Vachon MG. Dispersible phospholipid stabilized microparticles. WIPO WO030616A1, 2000.
48. McCulloch W, Parikh I, Colvin OM, Paro G. Development of I.V. busulfan (Spartaject- Busulfan). Annual Meeting of the European Group for the Bone Marrow Transplantation, Harrogate, UK, March 1994. 116.
49. Growdon JH et al. Lecithin can suppress tardive dyskinesia. N Engl J Med 1978;298:1029–1030.
50. Rathbone MJ et l. Modified-Release Drug Delivery Technology, New York, Marcel Dekker, 2003;171-172.
Received on 25.11.2009 Modified on 23.01.2010
Accepted on 19.02.2010 © RJPT All right reserved
Research J. Pharm. and Tech. 3(2): April- June 2010; Page 333-338