>>> Oral Formulation Optimization. Introduction. A Tiered Approach for Identifying Enabling Formulations

Similar documents
Biopharmaceutics Dosage form factors influencing bioavailability Lec:5

Copyright CSC Publishing

INTERNATIONAL JOURNAL OF INSTITUTIONAL PHARMACY AND LIFE SCIENCES

Accelerating Lipid-Based Drug Formulation Through Application of an Expert System

Robert G. Strickley, Ph.D. Senior Research Scientist II Gilead Sciences, Inc.

Aug 28 th, 2017 Pierre Daublain

Why and how does a pharmaceutical company take the risk to use novel excipients?

A FACTORIAL STUDY ON ENHANCEMENT OF SOLUBILITY AND DISSOLUTION RATE OF IBUPROFEN BY β CYCLODEXTRIN AND SOLUTOL HS15

Combining HME & Solubilization: Soluplus - The Solid Solution By: Hendrik Hardung, PhD; Dejan Djuric, PhD; and Shaukat Ali, PhD

Summary Chapter 8 CHAPTER 8. Summary. Page 173

formulation John K. Tillotson Abitec (SENDS)

Pharmaceutics I صيدالنيات 1. Unit 2 Route of Drug Administration

ADVANTAGES AND LIMITATIONS OF

Preparation and Characterization of Candesartan Cilexetil Solid Lipid Nanoparticulate Capsules

Determination of bioavailability

1. Gastric Emptying Time Anatomically, a swallowed drug rapidly reaches the stomach. Eventually, the stomach empties its content in the small

Soluplus The Solid Solution Opening New Doors in Solubilization.

STUDIES ON EFFECT OF BINDERS ON ETORICOXIB TABLET FORMULATIONS

1. If the MTC is 100 ng/ml and the MEC is 0.12 ng/ml, which of the following dosing regimen(s) are in the therapeutic window?

New formulas for successful drug delivery Hot-melt extrusion for enhanced solubility and bioavailability

Pharmaceutical Drug Development Consulting

7. SUMMARY, CONCLUSION AND RECOMMENDATIONS

Cremophor RH 40. Technical Information. For the pharmaceuticals industry

Functional Excipients for Suppository Applications

Enabling Lipid Formulations That Harness Supersaturation and Drug Absorption in the GI Tract

Journal of Chemical and Pharmaceutical Research, 2012, 4(8): Research Article

MODULE PHARMACOKINETICS WRITTEN SUMMARY

A FACTORIAL STUDY ON THE ENHANCEMENT OF DISSOLUTION RATE OF KETOPROFEN BY SOLID DISPERSION IN COMBINED CARRIERS

Preformulation Studies for Diltiazem in Self Emulsifying. Drug Delivery ABSTRACT. Mr Basant Behera* Asst Professor, IMT, Puri, India

ENHANCEMENT OF SOLUBILITY OF BICALUTAMIDE DRUG USING SOLID DISPERSION TECHNIQUE

MEDAK DIST. ANDHRA PRADESH STATE, INDIA. Research Article RECEIVED ON ACCEPTED ON

FORMULATION DEVELOPMENT OF RIVAROXABAN LIPID SOLID DISPERSION USING DIFFERENT TECHNIQUES FOR THE DISSOLUTION ENHANCEMENT

Available online at

Development and Validation of a New Uv Method for the Analysis of Rebamipide

International Journal of Research in Pharmaceutical and Nano Sciences Journal homepage:

Effect of Common Excipients on the Oral Drug Absorption of Biopharmaceutics Classification System Class 3 Drugs

Section 5.2: Pharmacokinetic properties

CAPMUL + CAPTEX + ACCONON = SEDDS

Dr. M.Mothilal Assistant professor

ISSN: X CODEN: IJPTFI Available Online through Research Article

Evaluation of combination drugs before the development of self-emulsifying drug delivery system

Delivery systems for nutraceuticals Enhanced bioavailability and improved functionality for lipophilic nutrients

A STUDY ON SUITABILITY OF NIMESULIDE-BETACYCLODEXTRIN COMPLEX IN ORAL AND TOPICAL DOSAGE FORMS

PHARMACOKINETIC STUDY OF DEXTROMETHORPHAN WITH URINARY EXCRETION

CHAPTER VI FACTORIAL STUDIES ON THE EFFECTS OF CYCLODEXTRINS AND SOLUTOL HS15 ON THE SOLUBILITY AND DISSOLUTION RATE OF EFAVIRENZ AND RITONAVIR

FORMULATION AND CHARACTERIZATION OF TELMISATAN SOLID DISPERSIONS

Development and validation of UV spectrophotometric estimation of lisinopril dihydrate in bulk and tablet dosage form using area under curve method

Public Assessment Report Scientific discussion. Anastrozole Bluefish 1 mg film-coated tablets (anastrozole) SE/H/781/01/DC

Tablet is a major category of solid dosage forms which are widely used worldwide. Extensive information is required to prepare tablets with good

Effect of a lipoidic excipient on the absorption profile of compound UK in dogs after oral administration.

BIOPHARMACEUTICS and CLINICAL PHARMACY

Chemate and Chowdary, IJPSR, 2012; Vol. 3(7): ISSN:

FACTORIAL STUDIES ON THE EFFECTS OF HYDROXY PROPYL β- CYCLODEXTRIN AND POLOXAMER 407 ON THE SOLUBILITY AND DISSOLUTION RATE OF BCS CLASS II DRUGS

Advantages and Limitations of In Vivo Predictive Dissolution (IPD) Systems

Biopharmaceutics Lecture-11 & 12. Pharmacokinetics of oral absorption

Asian Journal of Pharmacy and Life Science ISSN Vol. 2 (2), July-Sept,2012

THE INFLUENCE OF OILS AND SURFACTANTS ON THE FORMATION OF SELF-NANOEMULSIFYING DRUG DELIVERY SYSTEMS (SNEDDS) CONTAINING THERAPEUTIC PROTEIN

BASIC PHARMACOKINETICS

Excipient Interactions Relevant For BCS Biowaivers Peter Langguth

University of Sulaimani School of Pharmacy Dept. of Pharmaceutics Third level - Second semester

Development of Canagliflozin: Mechanistic Absorption Modeling During Late-Stage Formulation and Process Optimization

Synergistic Effect of PEG-400 and Cyclodextrin to Enhance Solubility of Progesterone Submitted: November 20, 2002; Accepted: January 9, 2003

Early BioPharm Risk Assessment in Discovery. Linette Ruston PCF9 17 th September 2010, Barcelona

SCIENTIFIC DISCUSSION

SELF-EMULSIFYING DRUG DELIVERY SYSTEM: A NOVEL APPROACH FOR ENHANCEMENT OF BIOAVAIBILITY

Public Assessment Report Scientific discussion. Nicorette Pepparmint (nicotine) SE/H/904/01/DC

TEPZZ 5459Z5A_T EP A1 (19) (11) EP A1 (12) EUROPEAN PATENT APPLICATION. (51) Int Cl.: A61K 9/10 ( ) A61K 9/107 (2006.

INTERNATIONAL JOURNAL OF PHARMACEUTICAL RESEARCH AND BIO-SCIENCE SELF EMULSIFYING DRUG DELIVERY SYSTEM (SEDDS): A REVIEW

Pharmacokinetics and allometric scaling of levormeloxifene, a selective oestrogen receptor modulator

Design and Optimization of Rivaroxaban Lipid Solid Dispersion for Dissolution Enhancement using Statistical Experimental Design. ), and Labrasol (X 3

SCIENTIFIC DISCUSSION. Darunavir

Research Article Derivative Spectrophotometric Method for Estimation of Metformin Hydrochloride in Bulk Drug and Dosage Form

Biopharmaceutics. Lec: 4

Suppository Chapter Content

Advantages and Limitations of In Vivo Predictive Dissolution (IPD) Systems

SUMMARY OF PRODUCT CHARACTERISTICS 2. QUALITATIVE AND QUANTITATIVE COMPOSITION

FDB FOOD AND DRUGS BOARD G H A N A GUIDELINES FOR CONDUCTING BIOEQUIVALENCE STUDIES

SCIENTIFIC DISCUSSION

LIQUID PREPARATIONS FOR ORAL USE. Final text for addition to The International Pharmacopoeia (November 2007)

Chapter 3. Need for Present Study

Validated UV Spectrophotometric Method Development And Stability Studies Of Acamprosate Calcium In Bulk And Tablet Dosage Form

Enhanced delivery methods for greater efficacy

SCIENTIFIC DISCUSSION. Lopinavir and Ritonavir 200 mg/50 mg Tablets * Name of the Finished Pharmaceutical Product:

Dealing with Uninvited Guests: Excipients. Mark Klang, MS, RPh, PhD, BSNSP Research Pharmacy Manager Memorial Sloan Kettering Cancer Center

USING PBPK MODELING TO SIMULATE THE DISPOSITION OF CANAGLIFLOZIN

FORMULATION AND EVALUATION OF VALSARTAN TABLETS EMPLOYING CYCLODEXTRIN-POLOXAMER 407-PVP K30 INCLUSION COMPLEXES

DEVELOPMENT OF UV SPECTROPHOTOMETRIC METHOD FOR THE ESTIMATION OF EZETIMIBE FROM TABLET FORMULATION

Ciprofloxacin Bluefish (Ciprofloxacin hydrochloride)

PK-UK Challenges and benefits of using PBPK to evaluate an IVIVC for drugs with nonideal solubility and/or permeability. Bath, November 2014

PHAR 7632 Chapter 7. Table Market and Share of Pharmaceuticals by ROA Data from Viswanathan, 2004

Lipid Based Drug Delivery Systems Focus Group

DERMAL APPLICATIONS. Topical and Transdermal. Penetration. Polarity. Captex. Acconon. Capmul MCM. Other EP/NF. Capmul. Captex 300.

PHA 5127 FINAL EXAM FALL On my honor, I have neither given nor received unauthorized aid in doing this assignment.

Preparation of 200 mg fenofibrate hard capsule with high dissolution profile with microparticle entrapped micelles technology

Industrial Pharmacy (3) Solutions as a dosage form. DR.Saad.M.YACOUB

Public Assessment Report. Scientific discussion. Celecoxib Apotex 100 mg and 200 mg, capsules, hard (celecoxib) NL/H/2760/ /DC

Full Length Original Review Article

Public Assessment Report Scientific discussion. Flucloxacillin Orion (flucloxacillin sodium) SE/H/981/03/DC

Public Assessment Report. EU worksharing project paediatric data. Valcyte. Valganciclovir

FROM SOLVENT TO AQUEOUS COATINGS. (Out of the Frying Pan...) Ralph E. Pondell. Coating Place, Inc. P.O. Box

Transcription:

Application Note #28-DMPK-3 >>> Oral Formulation Optimization Introduction Among the criteria required of compounds advancing from drug discovery programs, adequate systemic exposure (plasma concentrations or AUC) after oral dosing in preclinical models is one of the most important and sometimes most difficult to attain. Maximum oral bioavailability is desirable for both clinical and economic reasons. Low oral bioavailability is associated with high inter-subject variability of plasma concentrations, and therefore poor control of the drug s activities. And if bioavailability is low, much of the drug material is wasted, resulting in a certain economic disadvantage for costly drug substances. Incomplete oral bioavailability could be due to poor solubility, poor intestinal permeability, or presystemic metabolism. It is not at all unusual for the majority of compounds in a drug discovery program to have poor water solubility. For example, Lipinski estimated that about one third of all new Pfizer (Groton) compounds have solubility 5 μg/ml (1). Poor solubility is the one cause of incomplete oral bioavailability that can often be surmounted through formulation efforts. The optimization of oral bioavailability now usually begins early in the compound design and selection process, and in many pharmaceutical companies includes consideration of the influence of formulation on bioavailability. Productive drug discovery teams understand the limitations of poor water solubility, and know how to overcome these limitations. One of the most important studies used to compare active compounds within a discovery program is the initial pharmacokinetic study, which is typically performed in rats or mice. Because pharmacokinetic studies require both animal and analytical resources, and because the PK data are so critical for compound selection, these studies may become a bottleneck in the compound screening process. If a new compound in a series shows poor pharmacokinetic properties in the initial study, it is not likely to be re-tested or advanced. To avoid discarding developable compounds having the desired pharmacologic properties, it is prudent to give each compound the best chance for success in the initial PK study. For poorly water soluble compounds this may involve the use of a potentially enabling formulation. For maximum oral absorption of poorly soluble compounds it is necessary to solubilize the compound and keep it in solution in the gastrointestinal tract. We have used a tiered approach that includes in vitro solubility assays with minimum compound requirements to identify dosing vehicles that might be most useful for oral delivery of our Sponsors compounds. Here we describe this tiered approach to selecting dosing formulations for preclinical pharmacokinetic studies. We also provide examples of the application of this tiered approach for two poorly water soluble compounds already available as marketed drugs. A Tiered Approach for Identifying Enabling Formulations We believe that formulation efforts at the discovery stage should have the following objectives. To give developable compounds the best chance for success in the initial preclinical pharmacokinetic study. To use the simplest formulation as possible. The overall aim is to balance what it takes to get the compound absorbed, with consideration that the more heroic the dosing formulation is, the more difficult it may be for further development. If a compound is accepted for advancement to development based on PK results obtained using an unconventional formulation, there must be a feasible way to formulate that compound for safety studies and clinical trials.

A tiered formulation approach that can be used to evaluate poorly soluble compounds is summarized below. This table also lists some of the factors that will need to be considered later in development if this formulation approach is used. Table 1. Tier Formulation Components Factors that will be Important Downstream 1 2 3 4 Primarily aqueous solutions and suspensions with ph adjustment Primarily aqueous with solublizing agents - Cyclodextrins - Surfactants such as polysorbate 8, Cremophor, Soluene, vitamin E TPGS, poloxamers - Povidone - Dimethyl Isosorbide - Low concentrations of the solvents listed in Tier 3 Primarily cosolvent (+/- other solubilizers) - PEG 3, 4 - Propylene glycol - DMSO - DMA - Ethanol - N-methylpyrrolidone - Trimethylglycol Lipid based (+/- surfactants) - Oils - Medium chain glycerides - Labrafil - Labrasol - Gelucires Least formulation development issues Acceptability and cost of solubilizing agents Above plus Drug load and dose/tolerability limits of cosolvent Chemical and physical stability Is development of solid formulation possible? Above plus Formulation options may be restricted Cremophor (BASF) is a group of polyoxyl hydrogenated castor oil excipients. Solutol (BASF) is PEG15 hydroxystearate. Labrafil, Labrasol, and Gelucire (Gattefosse) are mixtures of glycerides and PEG esters. Tier 1 is the simplest formulations such as aqueous solutions and suspensions. If Tier 1 discovery formulations provide adequate oral absorption, adequate oral bioavailability should also be readily attainable with conventional solid dosage forms. Tier 2 formulations utilize an added level of complexity, the use of a solubilizing agent in a primarily aqueous dosing formulation. Compounds with adequate oral absorption using Tier 2 discovery formulations may also eventually achieve adequate oral bioavailability with conventional solid dosage forms. However, if Tier 2 dosing formulations are used in preclinical safety studies, the acceptability of the solubilizing agent will be an important consideration. Tier 3 discovery formulations are primarily non-aqueous. Some of the non-aqueous solvents that are acceptable for animal studies are listed in Table 1. Most of these, including DMSO, DMA, and ethanol, have limits for the percentage that can be dosed and the total amount that can be dosed. Therefore, Tier 3 formulations often are comprised of combinations of co-solvents. Compounds that attain adequate oral absorption with Tier 3 discovery dosing formulations may require more formulation development work than other compounds to achieve acceptable formulations for safety studies, clinical trials, and drug product. Tier 4 discovery formulations are comprised primarily of lipid components. These may be particularly effective in aiding the oral absorption of poorly water soluble compounds through their emulsifying activity. However, if only Tier 4 formulations are successful in achieving bioavailability, subsequent formulation development options may be limited. For each Tier of the discovery formulation approach there is a balance between the extent of oral absorption achievable and the possible limitations of the formulation at a later stage. The acceptability of any particular formulation approach will be different for each program, and possibly among different compounds within a program, and will vary from company to company.

To illustrate the use of the tiered formulation approach, we performed in vitro formulation evaluation and solubility studies to select dosing vehicles for rat pharmacokinetic studies with two poorly water soluble drugs, carbamazepine and itraconazole. We felt it would be useful to test some poorly soluble marketed drugs for which the feasibility of development is not in question. Some properties of these compounds are listed below. Table 2. Aqueous solubility Carbamazepine 158 µg/ml Itraconazole <3.5 µg/ml Molecular weight 236 76 clog P 2.38 5.66 Available products 2 mg tablets 1 mg capsule, 1 mg/ml solution (with hydroxypropyl-β-cyclodextrin) Absorption in humans (F) >7% 55% Methods Dosing formulations were selected through a preliminary examination of various potential formulations beginning with Tier 1 and progressing through Tiers 2 and 3, using pharmaceutically acceptable components and concentrations generally recognized as safe for preclinical use. The assessment of suitability for in vivo dosing was based on appearance (solubilization, viscosity) and conformance with QPS IACUC guidelines, as well as solubility after aqueous dilution. If the compound was not completely soluble in a vehicle used for dosing, aliquots of the dosing formulation were taken after filtration and analyzed to provide a measure of solubility in the dosing vehicle. Male Sprague Dawley rats weighing approximately 225-25 g were used in this study. Rats were obtained from Hilltop Research, and each had a jugular vein cannula (JVC) implanted for blood sample collections. Some rats were administered the test compound intravenously (IV), and these rats also each had a femoral vein cannula implanted for IV dose administration. There were 3 animals per dose group, and animals were fasted overnight before dosing until 4 hours post-dose. The doses used in this study were 1 mg/kg IV and 1 mg/kg PO. The dose volumes were 1 ml/kg for IV and PO, although itraconazole was also tested with some oral formulations using a dose volume of 1 ml/kg. The PO doses were administered by gavage. The IV doses were administered through the femoral vein cannula as a slow (<1 minute) bolus. Blood samples (.5 ml) were collected at the following times: PO: Pre-dose, 15, 3 min, 1, 2, 4, 6, 8 and 24 hr post-dose IV: Pre-dose, 5, 15, 3 min, 1, 2, 4, 6, 8 and 24 hr post-dose The whole blood was collected into tubes containing K 2 EDTA as an anticoagulant on wet ice and spun down within 6 min (at 4 C). Plasma (approximately.2 ml) was transferred to labeled tubes and stored at -2 C until analysis. Blood removed as specimens was replaced by blood transfusion using fresh blank rat blood. Sample analysis was performed using LC/MS/MS with internal standard after protein precipitation of plasma. Duplicate standard curves were run before and after samples. Calibration curves included double blank, single blank (Internal Standard only), and a minimum of 5 standards with LLOQ targeting approximately 1 ng/ml, prepared in blank rat plasma. Dosing formulation aliquots were analyzed after appropriate dilution in rat plasma. Non-compartmental pharmacokinetic analysis of the plasma concentrations vs. time data was performed using WinNonlin (Pharsight Corporation).

>>> Oral Formulation Optimization Results and Discussion Potential dosing vehicles were evaluated with the objectives of getting the compound in solution and keeping it in solution when diluted into an aqueous environment. Using the tiered approach, we started with the simplest dosing vehicles (Tier 1), and advanced through Tier 3 for these two test compounds. Tier 4 (lipid based) formulations were not evaluated for these compounds because it was already known that adequate absorption of these compounds should be attainable without the use of excipients in this category. One or more dosing formulation from each of Tiers 1, 2, and 3 was evaluated in vivo for each compound, so as to assess the impact of formulation on oral absorption. The in vitro assessment of various dosing formulations for each compound is summarized in Tables 1 and 2. The formulation assessment studies were performed using a few mg to prepare each potential formulation evaluated. Minimization of compound usage is very important at the discovery stage, when little compound is generally available. Compound usage can be further minimized by preparing a single concentrated stock solution in a solvent that can be dosed at a low concentration, such as dimethylsulfoxide (DMSO) or dimethylacetamide (DMA). To determine absolute oral bioavailability, each test compound was also administered IV. For carbamazepine, the IV dosing formulation was 1 mg/ml in 4% hydroxypropyl-β-cyclodextrin (HPbCD) in water. For itraconazole, the IV dosing formulation was 1 mg/ml in 1% dimethylacetamide, 6% PEG 4, 3% of 4% HPbCD in water. These dosing vehicles were identified based on the testing of oral formulations. Carbamazepine Pharmacokinetic results for carbamazepine are given in Figure 1 and the attached table. As summarized in Table 3, carbamazepine was not completely soluble in the Tier 1 formulations tested, but was soluble in the Tier 2 formulations. Carbamazepine remained in solution when Tier 2 and Tier 3 formulations were diluted 5X with water. The measured concentration in a filtrate of the 1% methylcellulose suspension formulation was.6 mg/ml, which is slightly lower than the reported aqueous solubility. The methylcellulose suspension formulation provided 5% oral bioavailability. When carbamazepine was dosed using the Tier 2 (4% HPbCD) or Tier 3 (DMA/PEG) dosing solutions, bioavailability was complete. Figure 2 shows individual plasma carbamazepine concentration vs. time profiles for the Tier 1 and 2 formulations. The suspension resulted in a delayed absorption profile, particularly in 2 of the 3 rats, compared with the solution formulation. Carbamazepine represents an example of a compound for which formulation influences bioavailability, but it has sufficient solubility such that acceptable properties in a discovery PK study would be obtained using the most basic formulation. The formulation and PK results we obtained are consistent with the hypothesis stating that getting the compound in solution and keeping it in solution can maximize oral bioavailability. Table 3. Carbamazepine Oral Formulation Evaluation Composition Observation 1 Observation 2 Pathforward Tier 1 1 mg/ml in 1% methylcellulose in water (Measured solubility =.6 mg/ml) 1 mg/ml in 1% methylcellulose Tier 2 1 mg/ml in 4% hydroxypropylβ-cyclodextrin (HPbCD) in water Soluble after sonication Soluble after diluting 5X with water Tier 3 1 mg/ml in 1% DMA / 9% PEG 4 Soluble Soluble after diluting 5X with water

>>> Oral Formulation Optimization Figure 1. Mean (+SD) Plasma concentration vs. time profiles and pharmacokinetic properties for carbamazepine in rats after 1 mg/kg IV or 1 mg/kg PO doses in various formulations. Figure 2. Plasma concentration vs. time profiles for carbamazepine in individual rats admimistered suspension (Rats #1, 2, 3) or HPbCD solution (Rats #4, 5, 6) formulations. 1 22 Plasma Carbamazepine (ng/ml) 1 1 1 IV 1 mg/kg Suspension HPbCD solution PEG solution Plasma Carbamazepine (ng/ml) 2 18 16 14 12 1 8 6 4 Rat #1 Rat #2 Rat #3 Rat #4 Rat #5 Rat #6 1 2 4 6 8 1 12 2 Time (hours) 2 4 6 8 Time (hours) 1 mg/kg PO C max (ng/ml) T max (h) AUC last t1/2 (h) AUC inf F (%) ND: Not determined Suspension HPbCD solution PEG solution Mean SD Mean SD Mean SD 332 195 174 315 1467 583 2.3 1.5.4.1.5. 1219 174 2561 725 2761 177 1.6 ND 1.5.5 1.5.5 1183 12 2573 721 2857 116 5 6 14 29 116 5 Itraconazole Itraconazole could be considered a more challenging compound to deliver orally, consistent with its lower solubility. The in vitro formulation assessment results shown in Table 4 indicate that it was not soluble in the Tier 1 or Tier 2 formulations tested, and a Tier 3 formulation was required for complete solubilization. Pharmacokinetic results for oral itraconazole are summarized in Figure 3 and its attached table. A methylcellulose suspension formulation (Tier 1) was dosed at both 1 mg/ml and 1 mg/ml concentrations for 1 mg/kg doses, but both resulted in variable and low oral absorption. The Tier 2, HPbCD formulation was dosed as a suspension and resulted in improvement of oral bioavailability to 15 ± 7%. The Tier 3 formulation improved oral bioavailability further to 56 ± 21%. It should be noted that this formulation resulted in itraconazole precipitation when diluted 5X with water. Further formulation optimization work might enable even greater itraconazole absorption in this preclinical model. Itraconazole represents an example of a compound for which formulation not only influences bioavailability, but could have been critical to the identification of this as a developable compound.

>>> Oral Formulation Optimization Table 4. Itraconazole Oral Formulation Evaluation Composition Observation 1 Observation 2 Pathforward Tier 1 1 mg/ml in 1% methylcellulose in water (Measured solubility <.1 mg/ml) 1 mg/ml in 1% methylcellulose (Measured solubility <.1 mg/ml) Dosed in vivo at 1 ml/kg Tier 2 1 mg/ml in 4% hydroxypropylβ-cyclodextrin (HPbCD) in water (Measured solubility =.25 mg/ml) 1 mg/ml in 4% hydroxypropylβ-cyclodextrin (HPbCD) in water 1 mg/ml in 1% dimethylacetamide (DMA) & 4% HPbCD in water 1 mg/ml in 1% dimethylacetamide (DMA) & 3% 3% Solutol HS15 in water Tier 3 1 mg/ml in 1% DMA / 89% PEG 4 / 1% polysorbate 8 Soluble Precipitates after diluting 5X with water Figure 3. Mean (+SD) Plasma concentration vs. time profiles and pharmacokinetic properties for itraconazole in rats after 1 mg/kg PO doses in various formulations. Plasma Itraconazole (ng/ml) 3 25 2 15 1 MC Suspension, 1 ml/kg MC Suspension, 1 ml/kg HPbCD suspension PEG solution Conclusions Here we propose a tiered approach for assessing discovery stage formulations and identifying the simplest formulations that can maximize oral absorption of poorly water soluble compounds. These formulation identification and pharmacokinetic studies provide an understanding of the formulation requirements for achieving oral absorption. This was illustrated with carbamazepine, which had reasonable bioavailablity with a simple methylcellulose suspension, and itraconazole, a more poorly soluble compound which required more complicated formulations to achieve oral bioavailability. 5 1 mg/kg PO C max (ng/ml) T max (h) AUC last t1/2 (h) AUC inf F (%) ND: Not determined 4 8 12 16 2 24 MC Suspension, 1 ml/kg Time (hours) MC Suspension, 1 ml/kg HPbCD suspension PEG solution Mean SD Mean SD Mean SD Mean SD 12 8 3 2 26 15 197 59 18. 1.4 2.9 4.4 4.7 3.1 3.3 2.3 165 128 2 2 32 126 183 657 1.2 ND ND ND 24.3 ND 4.6 ND 53 ND ND ND 59 34 1854 ND 5 4 <1 <1 15 7 56 21 References 1) C. Lipinski, Current Drug Discovery, April 21. 2) Zhou et al., J. Pharm. Sci. 96, 352 (27). 3) Goodman & Gilman s The Pharmacological Basis of Therapeutics (1996). Delaware Technology Park 3 Innovation Way, Suite 24 Newark, DE 19711 web www.qps-usa.com TEL (32) 369-561 Fax (32) 369-562