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

Similar documents
Approaches to the formulation of poorly soluble drugs

Working with Amorphous API s

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

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

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

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

Formulation Development of New Chemical Entities (NCEs) Dr. Mariella Artusi Pharmaceutical Development Dept. Monza

Oral Dosage Formulation Development

METHOCEL. TM Trademark of The Dow Chemical Company ( Dow ) or an affiliated company of Dow

Excipient Development at NCL

Pharmaceutical Sciences

Quantitation of amorphous content

PHARMACEUTICAL MANUFACTURING

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

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

Design and Dosage Form. Dr. Deny Susanti

Preface... iii Contents...vii Contributors...xv. 1 Introduction to Hot-Melt Extrusion, Continuous Manufacturing: Scale-up via Hot-Melt Extrusion...

Computation of Solubility parameters using Molecular. dynamics simulation

Development of paediatric formulations - points to consider

Practical Guide to Hot-Melt Extrusion: Continuous Manufacturing and Scale-up

Recent FDA Guidance For Industry; BCS Class 1 and 3 August 2015

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

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

How to Identify Critical Quality Attributes and Critical Process Parameters

Kollidon The Original Setting new standards in stability, purity and patient safety.

Solid Dispersions for Oral Administration: An Overview of the Methods for their Preparation

PHARMACEUTICAL AMORPHOUS ORGANIC MATERIALS CHARACTERIZATION BY USING THE DIFFERENTIAL SCANNING CALORIMETRY AND DYNAMIC MECHANICAL ANALYSIS

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

Outline CLINICALLY RELEVANT SPECIFICATIONS. ISPE Process Validation Conference September 2017 Bethesda, MD

Maximizing Roller Compaction Benefits with Proper Excipient Selection

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

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

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

Research Journal of Pharmaceutical, Biological and Chemical Sciences

Balancing the time, cost and risk of drug development. Christina Gustafsson, PhD Pharm, Formulation Scientist at Pharmaceutical Development, APL

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

Copyright. Siyuan Huang

A Basis For Understanding the Effects of Residual Water on the Mechanical Properties of Solids. George Zografi University of Wisconsin-Madison

INTERNATIONAL JOURNAL OF PHARMACEUTICAL RESEARCH AND BIO-SCIENCE

Pharma & Food Solutions. ETHOCEL One of the Few Water-Insoluble Polymers Approved for Global Pharmaceutical Applications

The main limitation of the classical basket- or paddletype

Drug Development and Incrementally Modified Drugs : Regulatory Perspective. American Association of Pharmaceutical Scientists October 27, 2015

NISSO HPC for Pharmaceutical Applications

Formulation and in vitro evaluation of bosentan osmatic controlled release tablets

Regulatory Assessment

Quality by Design Specifications for Solid Oral Dosage Forms: Multivariate Product and Process Monitoring for Managing Drug Quality Attributes

Roller Compaction: New trends, challenges and solutions

On-Demand Manufacturing of Pharmaceuticals

Roles And Applications of Cellulose Ether in Environmentally Friendly. Building Materials

Reflection paper on the dissolution specification for generic solid oral immediate release products with systemic action

TASTE-MASKING - Pharmaceutical Taste-Masking Technologies

INCOME December, 2008

Amorphous Content of Common Pharmaceutical Materials

Agilent TRS100 Raman. Quantitative Pharmaceutical Analysis System

Best Practices and Application of GMPs for Small Molecule Drugs in Early Development IQ Workshop, Feb 4-5, 2014, Washington, D.C.

AAPS Workshop on the Role of Dissolution in QbD and Drug Product Life Cycle: A Commentary

Formulation Development

Investigating Differences in Solubility Between Crystalline and Amorphous Forms of Pharmaceuticals

Pharma Silica Insights


QbD Applications for Lipid-Based Pharmaceutical Products. Alyn McNaughton

In vitro in vivo correlations (IVIVC) can be summarized

Application Note. Introduction. Analysis of crystal polymorphism by Raman Spectroscopy for Medicine Development

BCS Guidance and Biowaivers BCS Monographs

Application Note. Raman Spectroscopy Analysis of Crystalline Polymorphs for Pharmaceutical Development

2. Definition of Environmental Stress Cracking (ESC)

CONTRASOL : NOVEL NANOTECHNOLOGY FOR THE ENHANCED DEVELOPMENT OF INTRAVENOUS FORMULATIONS

Research Article. Hot Melt Extrusion: Development of an Amorphous Solid Dispersion for an Insoluble Drug from Mini-scale to Clinical Scale

Biowaiver Approaches for Generic Drug Products in the US: Case Studies

International Journal of Pharma and Bio Sciences DEVELOPMENT OF ACCELERATED STABILITY PROTOCOL FOR SILDENAFIL TABLETS A EUROPEAN PERSPECTIVE REVIEW

NOVEL DRUG DELIVERY SYSTEMS II

Bioavailability and Bioequivalence Studies

Pharmaceutical Sciences/Medicinal Chemistry, M.S.

A vapor (or gas) needs a completely enclosed container to have a definite volume; it will readily take on any shape imposed.

Recent Advances in Oral Granules and Bi-Layer Tablet Technologies, Solubility Enhancement Solutions, and Oral Disintegrating Tablet Applications

OVERVIEW OVERVIEW. At Particle Sciences, We Deliver, taking your API from concept to clinic. Contact us at to see how we can help.

The Raman effect, discovered in 1928 by C.V. Raman in his

capsugel.com Innovative Solutions Portfolio for Your Dosage Form Needs

Detection and Quantification of Amorphous Content in Pharmaceutical Materials

THE DISSOLUTION PROCEDURE: DEVELOPMENT AND VALIDATION

Pelagia Research Library

Formulation and Evaluation of Gastro retentive Bilayer Tablets- Glimepiride as Sustained Release and Lisinopril as Immediate Release

Quality by Design for ANDAs: An Example for Immediate-Release Dosage Forms

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS NOTE FOR GUIDANCE:

Scholars Research Library

Microcrystalline Cellulose, Colloidal Silicon Dioxide, Sodium Starch Glycolate, Sodium Stearyl Fumarate

Solubility enhancement of poorly soluble API using cellulose derivatives. 28th April 2014 Shilpa Mistry

In silico tools to study food-drug interactions, an Industry Perspective

TEST PROCEDURES AND ACCEPTANCE CRITERIA FOR NEW VETERINARY DRUG SUBSTANCES AND NEW MEDICINAL PRODUCTS: CHEMICAL SUBSTANCES

Insight into solid state properties during processing

Experimental design on product development

Pharmaceutical Sciences/Medicinal Chemistry, Ph.D.

Hot-melt extrusion with poorly soluble drugs

Pat Monitoring of Particles in API Manufacture and Formulation

Technological aspects of Encapsulation via Melt Extrusion Technology

INTERNATIONAL JOURNAL OF PHARMACY & LIFE SCIENCES

CERTIFICATE IN BIOPHARMACEUTICALS

Structure analysis from X-ray powder data using real-space methods and pair distribution function analysis

Chemical Activation of Low Calcium Fly Ash Part 1: Identification of Suitable Activators and their Dosage

Industry Perspective on Manufacturing in Early Development

Transcription:

2017 WHITE PAPER: Amorphous Solid Dispersions-One approach to improving Bioavailability Eric C Buxton Clinical Associate Professor University of Wisconsin Madison School of Pharmacy, Division of Pharmacy Professional Development

Page 1 Amorphous Solid Dispersions One approach to improving Bioavailability Though amorphous solid dispersions can add complexity to a drug development project, the difference they make can be the difference between bioavailable drug and failed project. During product development, the formulator s first task is to understand the properties of the API they are working with. This includes not just an understanding of the basic physicochemical properties, but the biopharmaceutical properties of the candidate as well. For BCS II drug candidates, a common product development approach is to utilize manufacturing processes that generate amorphous solid dispersions with a high degree of prolonged supersaturation. The choice of excipients for these formulations is A promising drug development project can be slowed or halted if the candidate compound has poor aqueous solubility and dissolution characteristics. critical not just to achieve drug delivery goals (bioavailability), but the excipients selected also need to be compatible with the selected manufacturing process. The excipients used and the specific formulation compositions selected also directly impact the stability of the disordered system and impact the packaging selection and dictate the environmental controls/handling requirements of the final product. Many small molecules cannot be crystallized in forms that are stable and suitable for formulation. Crystalline solids can be inadvertently rendered partially amorphous by processing (e.g. milling and drying) leading to instabilities. In addition, many crystalline APIs lack sufficient aqueous solubility to provide adequate oral bioavailability whereas the amorphous form can enhance dissolution characteristics. It is important to have adequate rate and extent of the oral absorption of solid API for bioavailability during various stages of the drug development process. For example, exposure is needed during early safety assessment, animal PK studies, First in Human (FIH) studies and for the final commercial product. While amorphous characteristics can complicate matters, they don t have mean that a promising project is halted. The utilization of disordered systems (amorphous solid dispersions) to optimize the delivery of poorly soluble (BCS II) drugs has become more commonplace due to an increase in challenging API properties. Drug candidates with properties similar to brick dust or grease balls are driving this need. To this end, our scientific understanding of disordered systems as well as the tools available to formulators has been increasing steadily over the past 20+ years. The number of new drug candidates with these issues has

Page 2 grown steadily over the past decades due in part to the use of high throughput and combinatorial screening tools during the discovery and selection phase. Currently there are a few drugs on the market with amorphous active pharmaceutical ingredients (APIs) including: Accolate (zafirlukast), Ceftin (cefuroxime axetil) Accupril (quinapril hydrochloride) and Viracept (nelfinavir mesylate). One of the development risks associated with amorphous products is their long term chemical stability is less than those of crystalline materials. Amorphous materials are typically less physically stable and tend to crystallize over time and under stress. Examples of stress are temperature and relative humidity, both of which can present storage issues for drugs over time. Crystalline drugs are typically more stable, but the strong interactions in the crystal lattice can lead to poor aqueous dissolution and poor oral bioavailability. Dissolution of solid crystalline material is a complex process. The crystal lattice of the solid needs to be disrupted to allow individual molecules to be solvated or hydrated. Thus, solvent/water hydrogen bonds must be broken to accommodate the solute molecules. This process can be represented by lattice energy, solvation energy, and cavitation energy. Solubility issues can be related to high lattice energy, poor solvation or a combination. There are methods to overcome aqueous dissolution limitations of crystals including chemical change (salt forms, ionizable prodrugs), formulation and processing (increasing specific surface area by George Zografi Emeritus Professor of Pharmaceutical Sciences University of Wisconsin Madison For over 45 years, George Zografi has carried out basic research to study the hydration of crystalline and amorphous solids as well as interfacial phenomena of surfactants and polymers as they relate to pharmaceutical systems. His pioneering studies in understanding the amorphous solid state, water-solid interactions, solid state stability, and wetting of solids have helped in the design and development of stable and functioning pharmaceutical dosage forms. His leadership in teaching and applying basic science to both preformulation and formulation activities has had profound effects on the drug development process as well as the field of pharmaceutics. His unique blend of scientific depth and rigor sprinkled with his human warmth and sensitivity have stimulated and promoted the research efforts of two generations of pharmaceutical scientists. particle size reduction or adding solubilizers), physical form selection (using more water soluble higher energy polymorphic crystalline forms and co crystals) and using amorphous solid forms. Amorphous materials lack the long range ordering of molecules typical for crystalline solids which gives rise to greater free energy and molecular mobility. Amorphous materials display a glass transition (Tg). A glass has

Page 3 the molecular conformation of a highly viscous liquid with the appearance of a solid due to very slow molecular motions and relaxation processes. These glassy materials represent the most energetic solid state of a material and it can be expected that these materials will offer useful solubilities and dissolution rates. (Figure 1) Figure 1: Hancock and Shamblin, Thermochimica Acta, 380 (2001) 95 These materials are inherently unstable, but combining the API with the proper excipients (that raise the Tg and thus decrease the molecular mobility or those that prevent nucleation and crystal growth) will stabilize the amorphous state during storage and prevent recrystallization during dissolution. The resulting amorphous solid dispersion can improve stability and dissolution. The amorphous drug is combined with a polymer that stabilizes the amorphous drug product resulting in better stability, higher apparent solubility and faster dissolution. An amorphous solid dispersion is formed by mixing the API with the polymer as a solid solution (miscible) usually through hydrogen bonding. Some of the major polymers used pharmaceutically to prepare amorphous dispersions include Polyvinyl pyrrolidone (PVP), Hydroxypropyl methylcellulose (HPMC), HPMC Phthalate, and Polyethylene glycol(peg). A stable dispersion is one with a Tg well above typical long term storage temperatures. Stability is also improved by storing the dispersions in a glassy state as free of absorbed water as is possible. Why would one want to deal with all of the potential instability issues with amorphous compounds and amorphous dispersions? Traditional alternatives for improving bioavailability include salt formation, decreasing particle size, using lipids for drug delivery, and co crystal generation. Salts are generally soluble if the counter ion can be hydrated. However, not all APIs have ionizable groups that can be used at biological ph values. In certain ph ranges the ionized forms of the salt may be metastable relative to the free forms. In addition, crystalline salt forms are generally more susceptible to hygroscopicity potentially leading to stability issues. Lipid drug delivery involves dissolving the API in a hydrophobic solvent that becomes available when encountering the GI environment in a supersaturated state. It offers the advantages of simple non aqueous solutions useful for early stages of

Page 4 drug development, the lipids can form nano sized particles, the lipids can take advantage of fat digestive processes, and they can be pelletized for later dosage form development. However, specialized equipment is required, crystallization may occur from solid lipid matrices, there can be chemical instability issues, and they are not as convenient and economical as tableting or capsules. Another method is to create co crystals which are molecular complexes containing the API and an additional non toxic molecular species in the same crystal structure. Advantages of co crystals for enhancing oral bioavailability include the intrinsic solid state physical and chemical stability of crystals relative to their amorphous form, and they are viable systems for enhancing oral bioavailability when supersaturation of API occurs in dissolution media and is maintained over therapeutically relevant timescales. However, there is a questions as to whether or not co crystals are new chemical entities (NCE) and patentable. Other potential disadvantages of co crystals include a requirement of stoichiometric amounts of co formers adding significantly to the required dose, the dissolution of co crystals and potential polymorphic transitions with hydrate formation when exposed to high relative humidity. The advantages of amorphous solid dispersions for enhanced oral bioavailability are: 1) Many crystalline drugs can be prepared in an amorphous form and be physically stabilized by an appropriate polymer and polymer composition; 2) Using formulation approaches, it is possible to maintain a sufficient amount of super saturation to enhance bioavailability in vivo, and 3) The currently used manufacturing methods (melt extrusion and spray drying) are adaptable from initially small samples for early studies to large scale manufacturing. As described earlier amorphous systems are thermodynamically unstable and subject to crystallization, have physical and chemical properties that change significantly at critical temperatures (Tg) and higher relative humidity. The choice of polymer and amounts cannot be accurately determined a priori, but there are basic principles that can be applied to speed up the formulation selection process. When pursuing an amorphous dispersion, it is incumbent upon the development team to screen for and characterize various API polymer combinations that provide miscibility, stability, and enhanced dissolution properties, while utilizing a suitable manufacturing process and packaging in order to produce a successful pharmaceutical product. An excellent chance to learn about amorphous dispersions from experts will be April 24 26, 2017 in Madison, Wisconsin. Understanding, Characterizing, and

Page 5 Developing Amorphous Materials and Amorphous Solid Dispersions features Professor George Zografi, a pioneer in this field, along with current industry experts. During this course, you will learn the various tools that can be used by formulator s tackling a challenging BCS II drug candidate. The tools discussed will include those specific to the goal of developing amorphous solid dispersions and include: characterization of the drug candidate, selection of excipients (formulating with a parachute ), identifying formulation compositions and understanding the challenges in early development. There are multiple examples in the literature that demonstrate how these concepts have been utilized and a few of these examples will also be discussed.