Approaches to the formulation of poorly soluble drugs

Similar documents
WHITE PAPER: Amorphous Solid Dispersions-One approach to improving Bioavailability

Model-based Technology Selection for Bioavailability Enhancement CPHI DAVID K. LYON, PH.D. OCTOBER 25, 2017

PHARMACEUTICAL PRINCIPLES OF SOLID DOSAGE FORMS

INTER CHANGEABILITY and EQUIVALENCE. Where we are and what we still have to determine!

ADVANTAGES OF MULTIPARTICULATES (PELLETS):

University of Sulaimani College of Pharmacy Dept. of Pharmaceutics 5 th stage Second Semester

Preformulation. By: Ass. Prof. Gamal Shazly

Soluplus. Technical Information. October _090801e-01/Page 1 of 8. = Registered trademark of BASF group. Pharma Ingredients & Services

FORMULATION STRATEGIES FOR LOW SOLUBLE DRUGS - AN OVERVIEW. René Holm, PhD Divisional Director Biologics and Pharmaceutical Science

USP s Perspective on Drug Product Performance Test

SOLID DISPERSION APPROACHES FOR SOLUBILITY ENHANCEMENT. Fig. 3. Fig 4

Oral Delivery of Drugs

Development of biopharmacy

PHYSICAL STABILITY OF SPRAY DRIED SOLID DISPERSIONS OF AMORPHOUS TOLFENAMIC ACID AND POLYVINYLPYRROLIDONE K-30

Biopharmaceutics Applications in Drug Development

Research Article Pharmaceutical Sciences

Solid dispersions as a formulation strategy for poorly soluble compounds. G. Van den Mooter University of Leuven, Belgium

DRUG SOLUBILITY EXECUTIVE SUMMARY

Nanoparticle Technology: Leveraging Rapid Dissolution to Improve Performance of Poorly Water-Soluble Drugs

Pharma Ingredients & Services. Soluplus. Technical Information. = Registered trademark of BASF group. July 2010 Supersedes issue dated May 2010

The Role of Polymer Excipients in Hot Melt Extrusion A Continuous Manufacturing Process

CHAPTER 1 PHARMACEUTICAL INDUSTRY

Accelerating development of enabled formulations for poorly soluble drugs

Pharma Silica Insights

BIOPHARMACEUTICS CLASSIFICATION SYSTEM-BASED BIOWAIVERS - M9

Received: ; Revised; Accepted:

Excipient Development at NCL

Cocrystals: A Regulatory Perspective. Scott L. Childs Renovo Research

Sung Je Lee 1 and David J. McClements 2

Design and Dosage Form. Dr. Deny Susanti

Particle Size Method Development and Validation in Support of NanoCrystal Colloidal Dispersion Formulation Characterization

Maximizing Roller Compaction Benefits with Proper Excipient Selection

The use of surrogates for dissolution testing for Immediate Release (IR) formulations, when is it feasible?

Investigation of Enhancement of Furosemide Solubilization with Cyclodextrins and a Novel Octenyl Succinate Anhydride Starch

Conducting Successful pre-ind Meetings to Reach FDA Concurrence for Sound 505(b)(2) Development

PHARMACEUTICAL MANUFACTURING

THE DISSOLUTION PROCEDURE: DEVELOPMENT AND VALIDATION

Solid Dispersion- A Review

University of Groningen. Combining the incompatible Drooge, Dirk Jan van

PACKAGING SOLUTIONS 68 George Buchinger Cora Helberg Cindy H. Dubin

Howida Kamal, Ph.D. Ass. Prof. of Pharmaceutics, Cairo University

At LATITUDE, we only do one thing, and we do it very well.

Deconvoluting Amorphous Solid Dispersion Dissolution

RapidFACT: Accelerated Formulation Development for Poorly Soluble Drugs and Modified Release Products

Microfluidics Reaction Technology (MRT) for Continuous, Bottom-Up Production of Drugs

APV Expert Workshop: The Preformulation Tool Box: The Key to Enter Successful Drug Development

Application of Quality by Design (QbD) in product development. James E. Polli September 16, 2015

Dissolution Enhancement of Active Pharmaceutical Ingredients in Eudragit E100 by Melt Extrusion Coupled with Supercritical Carbon Dioxide

GUIDANCE DOCUMENT Use of a Foreign-sourced Reference Product as a Canadian Reference Product

Abstract. Technical Aspects. Applying GastroPlus for Extensions of Biowaivers for BCS Class II Compounds 2

The research work highlights the development and evaluation of. bioavailability of drugs. The buccal route can bypass the first-pass

First UNGAP meeting Food-Drug Interactions Regulatory Aspects

Development of paediatric formulations - points to consider

Improving Oral Bioavailability by Solid Dispersions: An OSPHENA (Ospemifene) Case Study

Providing insight into pharmaceutical formulations

CONTRASOL : NOVEL NANOTECHNOLOGY FOR THE ENHANCED DEVELOPMENT OF INTRAVENOUS FORMULATIONS

NISSO HPC for Pharmaceutical Applications

Guidelines for Pharmaceutical Equivalence Requirements

Solubility Survival Guide. J u ly PharmTech.com

Supercritical Fluid (SCF) Drying of Pharmaceuticals Peter York Chief Scientist, CrystecPharma

Solid Phase transformation considerations during process development and manufacture of solid oral dosage forms A Review

The science of transforming an Active Pharmaceutical Ingredient (API) into a Drug Product (DP) in a specific dosage form.

Thermodynamics of Pharmaceutical Systems

7th Training School on Microencapsulation Strasbourg. Februar 2015 Textmasterformat in Mastervorlage eingeben

Predictability & Performance Through the Product Lifecycle Thought Provoking Perspectives

Overview of CDER Nanotechnology-related Drug Database

Edited by DENNIS DOUROUMIS. School of Science, JJniversity of Greenwich, UK. and ALFRED FAHR. Friedrich-Schiller University ofjena, Germany

How to Identify Critical Quality Attributes and Critical Process Parameters

USE OF HARD CAPSULES AS CONTAINER IN LIPID BASED FORMULATIONS. CAPSULE FILLING TECHNOLOGY. December, 2017

LYOPHILIZATION. Introduction to Freeze-Drying Third of Four Lectures. Freezing, Annealing, Primary, and Secondary Drying. J. Jeff Schwegman, Ph.D.

CRISTALIZATION UNIT YUSRON SUGIARTO

ASSESSING THE DRUG RELEASE FROM NANOFORMULATIONS WHEN, WHY AND HOW?

ENTEK Membranes LLC Lebanon, Oregon USA

QUALITY OF PROLONGED RELEASE ORAL SOLID DOSAGE FORMS

From Candidate to Clinic: Strategies to Select, Assess, Formulate, and Deliver the Right Drug Candidate in the Early Phase

Chimica Farmaceutica. Pharmacokinetics and related topics

BCS, Biowaivers and Dissolution Test Methodologies

Microfluidizer Processors:

CHALLENGES & OPPORTUNITIES OF ICHQ8 (PHARMACEUTICAL DEVELOPMENT) AN INDUSTRY PERSPECTIVE

DEVELOPMENT PHARMACEUTICS AND PROCESS VALIDATION

Amorphous drugs and drug delivery systems

Injection Moulding as a One-Stop-Production to Produce Pharmaceutical Dosage Forms

A practical computational tool to predict formulation and process variables during the development of spray-dried amorphous solid dispersions.

Solubility of Small-molecule Drugs into Polymer Excipients in Hot Melt Extruded Dosage Forms

Porous Inorganic Excipients as Carriers for Amorphous Solid Dispersions

USE OF SPHERICAL TECHNOLOGIES

Biowaivers and Harmonization Guidelines for Class I and 3 Drugs: Biowaiver Case Studies

Meeting Solid Dose Formulation Challenges

Who we are? Dissolution testing of modified release forms. Dissolution testing of immediate/ modified release forms

Vol - 4, Issue - 2, Apr 2013 ISSN: Dhumal et al PHARMA SCIENCE MONITOR

Chapter 1. Introduction

Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, PR-00936, United States

7. SURFACE TENSION AND DETERMINATION

Kollidon SR: A polyvinyl acetate based excipient for DCsustained-release

PROFESSOR VINESS PILLAY & DR. YAHYA E. CHOONARA Department of Pharmacy and Pharmacology at the University of the Witwatersrand

Adare Pharmaceuticals. A partnership that adds value to your products

Computation of Solubility parameters using Molecular. dynamics simulation

Formulation Development & CTM Manufacturing Services

Indian Journal of Pharmaceutical and Biological Research (IJPBR)

TASTE MASKING WITH COATINGS. Coating Technology 96 October 8-9, Presented by Ralph E. Pondell Corporate Secretary

Transcription:

1 Approaches to the formulation of poorly soluble drugs R. Christian Moreton, Ph.D FinnBrit Consulting ExcipientFest 2013, April 30 May 01, 2013, Baltimore, MD 2 Disclaimer The views expressed in this presentation are my own, and should not be interpreted taken as the official view of any organization with which I am associated. In the interests of time, this presentation will only consider oral drug delivery. Some of the approaches presented may apply to other routes of administration; however there may also be other approaches that will work for other routes of administration. This will be a very brief overview to show what techniques are available.

3 Presentation Outline Introduction Poorly soluble drugs Absorption from the gastrointestinal tract Dissolution models Formulation approaches for poorly soluble compounds Solubility-based methods Surface area-based methods Lipid-based systems Potential impact for excipient manufacturers Summary 4 Poorly soluble drugs Depending on the therapeutic area as many as 80% of new drugs are classified as poorly soluble. Poorly soluble is defined in the Biopharmaceutical Classification System (BCS) as not being soluble in 250 ml of aqueous media over the range ph1 ph7.5. From my personal experience, poorly soluble means that it is soluble at <1 mg/ml in dilute HCl. There are differences between solubilities of >500 mcg/ml and <10 mcg/ml with respect to the types of formulation approach we can use. Issues for clinical and commercial formulations. Issues for preclinical safety formulations.

5 Biopharmaceutical Classification System High II I Permeability Low IV III Low High Solubility 6 Absorption from the gastrointestinal tract Passive diffusion Paracellular Transcellular Carrier mediated Facilitated diffusion Active transport Efflux 1 2 3 5 4 1 Transcellular 2 Paracellular 3 Carrier mediated 4 Transcytosis 5 Efflux

7 Dissolution models Noyes Whitney Nernst Brunner Where dq/dt = dissolution rate k = mass transfer coefficient C s = solubility C b = bulk solution concentration D = diffusion coefficient A = solid surface area h = diffusion layer thickness Thus we have three possible ways to influence the dissolution: Solubility Surface area Diffusion layer thickness [Within the confines of the gastrointestinal tract, we do not have much control over diffusion layer thickness.] When we talk about solubility, it can be either equilibrium solubility, or dissolution rate, or both. Increasing the rate of dissolution may lead to supersaturation. The extent of supersaturation (concentration and time) are both important. 8 Dissolution models (contd.) Ostwald Freundlich Kelvin 2 2 Where S = drug solubility at temperature T S 0 = equilibrium solubility (r = ) r = particle radius M = molecular wt. of solute ν = molar volume γ = interfacial surface tension ρ = true density of solute From this equation we see that dissolution increases as the particle radius decreases. This effect only really becomes apparent below 1 2 μm, and especially below 200 nm, i.e. in the nano range.

9 Formulation methods Solubility We are trying to increase the concentration gradient across the diffusion layer: Alternative salt forms Metastable crystalline polymorphic forms Co-crystals Complex formation Co-solvents Solubilization by surfactants Amorphous forms 10 Alternative salt forms There are many different potential salts from both weak acids and weak bases. Can enhance solubility, and thus dissolution and bioavailability. Increase in bioavailability may be 1.5 2.5 fold. Not really suitable for very insoluble drugs, i.e. S 0 << 1mg/mL. May also increase stability of the API e.g. amlodipine maleate amlodipine besylate

11 Metastable polymorphs Inherently unstable. They will tend to revert to the most stable (least soluble form) Some are sufficiently stable to be viable. Moisture may be an issue for stability. Increase in bioavailability may be 1.5 2.5 fold. Not really suitable for very insoluble drugs, i.e. S 0 << 1mg/mL. 12 Co crystals These are not salts; it is not necessarily an ionic association. There may be hydrophilic (hydrogen bonding) and hydrophobic interactions. Each API will be different, and what may work with one molecule will not work with another closely related molecule. Can provide a 2 4 fold increase in bioavailability. Stability can be an issue. Sometimes other components can be added to stabilize the co-crystals.

13 Complex formation Complexes based on shielding of the hydrophobic portions of the molecule: e.g. cyclodextrin complexation The complex must release the drug molecule in the gastrointestinal tract for the drug to be absorbed. 14 Co solvents Water miscible co-solvents The final formulation may be a solution, partial solution/suspension, and either liquid or solid. Solid solutions may be preferred since they are likely to be more stable to moisture. Liquid co-solvent solutions can crystallize on storage in e.g. soft gelatin capsules due to moisture uptake from the capsule shell. A partial solution/suspension can give extended release in vivo. Examples include: Gris-PEG : Griseofulvin in PEG 400/PEG 8000 Aprical : Nifedipine in PEG 300/PEG 6000

15 Solubilization by surfactants Surfactants have the ability to form micelles in aqueous media. The drug molecule is able to be solubilized in the hydrophobic inner core of the micelle. There may be problems with the amount of surfactant required (e.g. laxative effects). The micelles must release the drug molecule for it to be absorbed. 16 Amorphous forms Amorphous forms of drug molecules are high energy forms. Since there is no crystal lattice structure (no long range order), the energy barrier to dissolution is much reduced. Inherently unstable, and will tend to revert to the most stable (least soluble form) Amorphous forms most often need to be stabilized Solid solutions Polymer dispersions Can give up to a 10 15 fold increase in bioavailability. Methods of manufacture include: Spray drying Hot-melt extrusion Conversion to the amorphous form can also be achieved by milling (e.g. ball milling)

17 Formulation methods Surface area An increase in specific surface of the bulk active drug (API), can aid dissolution: Milling Micronization Nanomilling Nanocrystals The lower the equilibrium solubility of the drug, the smaller the particle size needed to get adequate drug dissolution, and thus absorption. 18 Milling methods Milling is the breaking down large particles to smaller particles. At the same time the total surface area is increased. There are several milling methods, but they can be grouped as either wet or dry methods. For particle size reduction beyond what can be achieved air-jet milling (micronization), wet milling is necessary.

19 Dry milling There are several different types or dry milling equipment. The most common in the pharmaceutical industry are: Comminuting mills (e.g. Fitzmill, Comill, etc.) Pin mills (either with one rotary head or contra-rotating heads) Fluid energy mills The more energy we put into the milling operation, the finer the resulting particles. However, there is a lower limit for dry milling since powder particles become more cohesive the smaller they get. Using dry milling methods we can get down to a median particle size of ca. 1 10 m (e.g. using fluid energy milling), depending on the material being milled. 20 Wet milling As the name implies, this is milling where the powder to be milled is suspended in a liquid. Wet milling methods include: Ball milling Bead milling Colloid milling Microfluidization Easier to handle very fine powders as a suspension: Reduced dust. Reduced agglomeration/crystallization e.g. through including crystallization and agglomeration inhibitors dissolved in the liquid. For every material and type of milling there is a particle size below which it will not be possible to obtain any further size reduction.

21 Nanocrystallization Refers to crystallization under conditions of very high shear to produce nano-sized crystals: Drug and stabilizing polymer in solution, and an antisolvent are fed under high pressure into the crystallization chamber and caused to impinge directly. The conditions of very high shear creates very nanocrystals, and the presence of the stabilizing polymer, which adsorbs onto the nanocrystal surface, prevents agglomeration and crystal growth. Particle sizes <100 nm are claimed 22 What is the target particle size? It depends on the drug and its solubility: Griseofulvin: Aqueous solubility: 0.1 1 mg/ml Critical particle size: <ca. 10 m Digoxin: Aqueous solubility: ca. 0.01 0.02 mg/ml Critical particle size: <5 m Investigational compound: Aqueous solubility: <50 ng/ml Critical particle size: <300 nm

23 Mesoporous particles Highly porous particles with nano-sized pores that are able to take up the drug from a solvent solution and then release it in the body after administration. Particles typically prepared from amorphous silica. Other materials may be possible. The drug-laden particles can be further formulated for encapsulation or compressed into tablets. 24 Formulation methods Lipid based Lipid-based systems are alternatives to the non-lipid-based formulations; however, drug solubility and surface area still apply. Self-emulsifying systems (SEDDS) Self-microemulsifying systems (SMEDDS) Lipid solutions Solid lipid nanoparticles (SLNs)

25 SEDDS and SMEDDS Both types of formulations are combinations of the API with a mixture of oils and surfactants which emulsify on addition to an aqueous phase. The difference between the two types concerns the particle size of the discrete phase. SEDDS have more coarse droplets and the emulsions are opaque when sufficiently concentrated. SMEDDS have much finer droplets and the resulting microemulsion is clear/translucent, even when concentrated 26 Sandimmun vs. Neoral Sandimmun a SEDDS; Neoral a SMEDDS Neoral is less dependent on bile salts for absorption. Neoral gives better, more linear, and less variable bioavailability Choc, MG, Int J Dermatol, (1997) 36 (Suppl 1), 1 6

27 Lipid solutions Can be used with some drugs. The lipid should be a liquid at body temperature. Natural fixed oils (vegetable oils) are preferred. Can be a useful formulation for short term preclinical safety studies. Has been used to administer oil-soluble vitamins. 28 Solid lipid nanoparticles Reports from the literature have shown improved bioavailability using this formulation approach. SLNs are prepared using high pressure homogenization techniques. Median particle sizes <300 nm have been reported.

29 Potential drawbacks for all methods Stability: Physical, Chemical. Drug loading: Typically <50% Potential problem for high-dose drugs. Problems for safety studies: Volume of material to be administered. Dose of excipients may be too high. High doses of surfactants is a problem. 30 Implications for excipients For poorly soluble compounds we need to focus on getting the drug into a form from which it can be absorbed. This is in contrast to more soluble drugs where we were able to focus on what was needed to formulate the tablet or capsule. Once we have the drug in a suitable form for absorption, we can then focus on what else is needed to produce the formulation to be administered to patients. We need better understanding of how to increase the drug loading of these formulations; particularly for preclinical safety studies. We need new excipients/excipient combinations capable of providing higher drug loadings and more stable formulations. Note: we will still need conventional excipients to manufacture the final product.

31 Summary Very poorly water-soluble compounds are going to be a big part of the future for pharmaceutical formulation. There are methods available, and under development, that can provide a means to administer such drugs to patients and achieve a reasonable bioavailability. None of these methods are fully understood. We need better understanding of how these methods work. We need better excipients/excipient combinations to provide more stable formulations with higher drug loadings. Preclinical safety studies will continue to present issues when working with very poorly soluble drugs. 32 Thank you! Any questions?