european Industrial Pharmacy Issue 19 (December 2013)

Page 1

european INDUSTRIAL

PHARMACY features 4

SCALE-UP OF SPRAY DRIED AMORPHOUS SOLID DISPERSIONS Technologies capable of addressing solubility issues are needed in light of the many insoluble drugs in development. The authors discuss the use of stabilised amorphous solid dispersions prepared using pharmaceutical spray drying. by José Luís Santos, Paula Cordeiro, Márcio Temtem

9

THE INNOVATIVE MEDICINES INITIATIVE OrBiTo PROJECT – DEVELOPING THE NEXT GENERATION OF PREDICTIVE TOOLS FOR ORAL BIOPHARMACEUTICS The Innovative Medicines Initiative OrBiTo project is a collaboration between pharma, academia and specialist technology companies to enhance our understanding of absorption of orally-administered drugs from the gastrointestinal tract and apply this knowledge to predict the performance of oral formulations in patients. by Mark McAllister, Bertil Abrahamsson, Hans Lennernäs

12

FORMULATION TECHNOLOGY ENABLES THE DELIVERY OF HIV MEDICINES Innovative formulation technologies have been developed to overcome drug delivery challenges of treatments of HIV infection. This article describes problems of solubility, pharmacokinetics and physical material properties along with their resolution in a series of case examples. by Peter Timmins, Jonathan Brown

18

TAKING STEPS TO IMPLEMENT THE FALSIFIED MEDICINES DIRECTIVE AND COMBAT COUNTERFEITING IN EUROPE – THE EUROPEAN STAKEHOLDER MODEL Falsified medicines present a public health threat at global level, with counterfeit goods threatening the health of patients in countries around the world. This article discusses measures to fight counterfeiting. by Richard Bergström

regulars 3 20 21 23

EDITORIAL COMMENT REGULATORY REVIEW NEWS FROM THE EIPG EVENTS

ISSUE 19 • DECEMBER 2013 www.industrialpharmacy.eu www.eipg.eu


associate editors

european

INDUSTRIAL

PHARMACY

Belgium: Philippe Bollen

Issue 19 December 2013 Bulgaria: Valentina Belcheva

ISSN 1759-202X

Czech Republic: Ales Franc

MANAGING EDITOR Sue Briggs

Denmark: Marie Fog

PRODUCTION Dave Johnson

Finland: Anni Svala

SUBSCRIPTIONS Jill Monk

France: Jean-Pierre Paccioni Germany: Armin Hoffmann Great Britain: Shilpa Gohil, Janet Halliday Greece: Ioannis Nikolakakis

EDITORIAL BOARD Michael Anisfeld Claude Farrugia Michael Gamlen Linda Hakes John Jolley European Industrial Pharmacy is published four times a year by: Euromed Communications Passfield Business Centre, Lynchborough Road, Passfield, Liphook, Hampshire GU30 7SB

Hungary: Sylvia Marton Ireland: Anna O’Mahony Italy: Piero Iamartino Latvia: Inta Saprovska, Anita Senberga Malta: Claude Farrugia

Tel: +44 (0)1428 752222 Fax: +44 (0)1428 752223 Email: info@euromedcommunications.com www.industrialpharmacy.eu Annual subscription rate £90

Netherlands: Amon Wafelman

european INDUSTRIAL PHARMACY

Norway: Wenche Gordon

discussion group: www.pharmweb.net/gmp.html

Portugal: Nuno Moreira Views expressed in European Industrial Pharmacy are those of the contributors and not necessarily endorsed by the Publisher, Editor, Editorial Board, or by our corporate sponsors who accept no liability for the consequences of any inaccurate or misleading information

Spain: Beatriz Artalejo Sweden: Marianne Andersson Switzerland: Valter Gianesello

© 2013 Euromed Communications

european INDUSTRIAL PHARMACY is the official publication of the European Industrial Pharmacists Group (Groupement des Pharmaciens de l’Industrie en Europe) www.eipg.eu

2

Cover photo: HIV virus (see Formulation technology enables the delivery of HIV medicines on page 12)

european INDUSTRIAL PHARMACY December 2013

• Issue 19


editorial Dear Colleagues Welcome to the December edition of the European Industrial Pharmacy journal. Over the past 3 months, a number of actions have been undertaken by members of the Bureau and, in particular, I would like to emphasise the revamping of the website. This has been possible because of the tireless efforts of Dr Claude Farrugia, Vice-President, Communications, whom I would like to thank warmly for his dedication and support in this respect. Rather than describing the new features and advantages of our website, I would like to invite you to pay it a visit at the following address: http://eipg.eu. The new website has been designed to raise the profile of the EIPG and its communications with its members and with pharmaceutical industry pharmacists in general, irrespective of whether they are employed within the European Union or outside it. I consider it essential for the work of the EIPG – position papers, publications, presentations,

guides, codes, contributions to meetings with other European professional organisations and institutions – to be made rapidly available to all. By way of an example, I would like to invite you to check out our position papers: 'EIPG Feedback on Revision to Annex 16' and 'Joint call for actions on medicines shortages from European pharmacist organisations'. I also consider it essential that visitors to our website become frequent visitors and would like to advise regular use of the site as a source of news and information for industrial pharmacists. Lastly, in the context of strengthening the EIPG's partnerships with key stakeholders, I have the pleasure of announcing that EIPG has been accepted as an observer in the USP Convention, since several opportunities for close collaboration with the USP have been identified. May I conclude by wishing you a very Merry Christmas and a Happy New Year. Jean-Pierre Paccioni EIPG President

Clinical Research Manual Editors: David Luscombe and Peter D Stonier 20 Chapters over 600+ pages £190.00

NEW EDITION ical Clinearch Resanual M

“a real manual – user-friendly, printed and edited in a style that induces the reader to continual consultation” Applied Clinical trials

rs Edito mbe usco L r id Dav tonie rDS Pete

2013

A practical, international handbook for all those working in clinical research. In over 600 pages and 20 chapters, the twentyfive different authors and experts cover the the entire drug development programme from Drug Discovery through to Postmarketing Surveillance. The Editors have a wealth of experience of teaching and practice of clinical research, and have selected and reviewed each chapter to ensure completeness and relevance.

io Edit

n

“All chapters are well written and some of them are really excellent. A lot of useful information can be found both by beginners and experts” Arzneim-Forsch/Drug Res

For details and to order please visit the Euromed website:

www.euromedcommunications.com

european INDUSTRIAL PHARMACY December 2013

• Issue 19

3


SCALE-UP OF SPRAY DRIED AMORPHOUS SOLID DISPERSIONS by José Luís Santos, Paula Cordeiro, Márcio Temtem

W

ith an increasing number of relatively insoluble drugs in development today, there is a need to develop and use platform technologies capable of addressing solubility issues. Among the different alternatives, the use of stabilised amorphous solid dispersions is becoming increasingly popular, with pharmaceutical spray drying being one of the key technologies used in their preparation. In spray dried dispersions (SDD), the active pharmaceutical ingredient (API) is molecularly dispersed in a polymeric matrix. The polymer is used to stabilise the amorphous, metastable form of the drug and sustain supersaturation of the API in solution/biological fluids, thereby increasing bioavailability. José Luís Santos is a Drug Product Development Scientist at Hovione. He joined the company in 2011 and has worked on many projects involving development of spray dried dispersions, manufacture of micro and nanoparticles using integrated milling and membrane technologies, and implementation of scale-up and modelling methodologies. (email: jlsantos@hovione.com) Paula Cordeiro is a Drug Product Development Scientist at Hovione. She joined the company in 2011 and has worked on many projects involving development of spray drying processes with the use of scale-up and modelling methodologies, spray congealing processes and milling technologies. Márcio Temtem is Group Leader and Senior Scientist in the Drug Product Development team at Hovione. He joined the company in 2008 and has been technically involved in development of spray drying processes, amorphous solid dispersions, milling technologies, biodegradable release systems and oral final dosage forms.

Spray drying is a well-established unit operation, mostly owing to intense process optimisation in the food industry where profit margins are tighter. In pharmaceutical spray drying, powder properties (e.g. particle size and bulk density) are typically considered critical quality attributes and because of this a good process understanding is fundamental to enabling a seamless transfer between spray drying units/scales with the smallest number of trials and a minimal use of API. Dobry et al.1 proposed a scale-up methodology for developing SDD formulations with minimum API. Their approach is based on a

4

scientific understanding of the spray drying process and critical material attributes that need to be considered: amorphous stability, chemical stability to high temperatures, thermodynamic spaces of the different spray dryer units, atomisation assessment and droplet size measurement, drying kinetics in the drying chamber and experimental confirmatory runs. Such work, however, does not include detailed information on the impact of raw materials and process conditions on the SDD critical quality attributes. In a review paper by Vehring2, a theoretical framework was established for determining the

mechanisms of particle formation. Despite not considering local interactions with the drying gas flow, such simple approach can provide an understanding of what can be the structure and morphology of the spray dried particles. Nevertheless, due to the characteristics of the API, each formulation is unique, posing great challenges in the accurate prediction of final powder properties. A recent contribution by Paudel et al.3 reviews the state-ofthe-art, from an academic perspective, including several examples of the impact of raw materials and process conditions on the SDD properties. This is, however, limited to laboratory- and pilot-scale equipment, and does not include industry-relevant case studies – the authors are clear in stating that the literature is scarce in that respect. A recent work from Ullum et al. proposed the use of computational fluid dynamics (CFD) through the investigation of the flow profile of the drying gas in a spray dryer coupled with droplet drying4. Additionally, recent advances in the modelling of spray drying by CFD have been discussed5, including its use for pharmaceutical applications6. The complexity of the spray drying process, where phenomena occur on vastly different time and spatial scales, suggests that the computational power still needs to evolve substantially before the particle formation process can be adequately predicted within a plant-scale CFD simulation. In this current paper, the authors aim to discuss the different challenges and perspectives of the scale-up of SDDs based on their experience with a large number of projects spanning from the laboratory up to the large pharmaceutical commercial scale. It is the authors’ intention to share industry-relevant information and opinion on what needs to be considered in the scale-up of SDDs, hoping that it can contribute to improve the scientific and technical understanding of the relation of SDD quality attributes with both raw material attributes and process conditions.

european INDUSTRIAL PHARMACY December 2013

• Issue 19


SCALE-UP OF SPRAY DRIED AMORPHOUS SOLID DISPERSIONS

continued

Stability of amorphous solid dispersions

One of the most important attributes of an amorphous solid dispersion is its glass transition temperature (Tg), which is intrinsically related to the API molecular mobility and is one of the characteristics that dictates whether an SDD formulation is stable enough not to change significantly over the shelf-life of the product. To reduce the molecular mobility, a typical rule of thumb used during formulation development is to target a single Tg (true solid solution) of at least 50ºC higher than room temperature7, i.e. a single Tg above 75ºC is ideal from a product shelf-life perspective. Phase separation, as shown by multiple Tgs, is typically associated with the presence of clusters (API or polymerrich phases) in the matrix and, therefore, the material presents a higher potential for recrystallisation. Both polymer/excipients and API contribute to the Tg of the formulation, which can be estimated by simple equations that weight the individual Tgs of the API, polymer and solvent, e.g. the Fox or Gordon–Taylor equations8. Deviations to these predictions are typically attributed to interactions between the different ingredients and should be considered as positive events from a stability perspective. The inherent use of solvents in spray drying processes also contributes to lowering the Tg of the SDD through a plasticisation effect attributed to the solvent and this should be considered during process development and scale-up. In addition to the supersaturation/ bioavailability considerations that are not covered in this paper, laboratory-scale SDD development primarily focuses on achieving high and single glass transition materials, so that the formulation is stable and the process can be developed with a good yield. To prepare the scaleup and account for the operation in closed loop units (i.e. with nitrogen recycle), a few trials are executed with varying drying conditions to generate SDD materials with distinct residual solvent contents. The Tg of

Figure 1: Tg and relative saturation of the solvent in the drying gas (RS_out) as a function of residual solvent content in the SDD (circles: laboratory scale spray dryer; squares: large scale spray dryer).

those materials is then assessed by modulated Differential Scanning Calorimetry (mDSC) in a closed pan to assess the plasticisation effect of the solvent. A useful illustration of these results is presented in Figure 1, where the drying conditions, expressed as the relative saturation of the solvent in the drying gas (RS_out), and Tg are plotted as a function of the residual solvent content in the SDD. The plasticisation effect is scaleindependent and is used to support the selection of the most suitable drying condition. Included in Figure 1 are laboratory and large-scale trials that can be taken as representative of the majority of SDD formulations. Conversely, the relation between relative saturation in the gas stream and powder solvent content is scale-dependent as it is related to residence time and droplet/particle size. In particular cases, the trends of both spray drying scales are coincident as illustrated in Figure 1, which indicates that the data generated on amorphous stability in the laboratory with a small quantity of API can aid the preparation of the scale-up for commercially ready spray drying units. Figure 2 illustrates one such case, where the material from the larger-scale spray dryer matched perfectly the Tg and X-ray powder diffraction (XRPD)

european INDUSTRIAL PHARMACY December 2013

• Issue 19

profile of the SDD produced in the laboratory. Furthermore, the data from laboratory-scale trials can generally be regarded as a worstcase scenario since the particles’ residence time in the drying chamber is lower than in larger-scale spray dryers, causing the solvent content of laboratory-generated SDD to be typically higher. The secondary drying of wet SDD materials is another common operation in the manufacturing of amorphous solid dispersions. To achieve the International Conference on Harmonization limits for the residual solvents, tray dryers, doublecone dryers or static agitated dryers are most widely used. The same set of data shown in Figure 1 can also be used in the optimisation and development of a secondary drying strategy. Figure 3 shows secondary drying data, where the Tg of the powder is increasing with a decrease in residual solvent content. A conservative approach is to always use drying temperatures below the SDD Tg, which can be adjusted incrementally during the operation as illustrated in Figure 3 to decrease the secondary drying cycle time. SDD downstream processability and performance

The downstream performance of a spray dried material may be

5


SCALE-UP OF SPRAY DRIED AMORPHOUS SOLID DISPERSIONS

continued

Figure 2: DSC (top left/right) and XRPD (left) for SDD produced at both scales (blue lines: laboratory; red lines: large scale).

discussed from two different perspectives: processability and product performance. Typical downstream operations include blending, roller compaction and tableting. The ability for an SDD material to flow and be processed in the downstream equipment with no major operational difficulties is closely related to powder properties, namely particle size, density and cohesive–adhesive balance between the ingredients9. Rule-of-thumb strategies for improving flow indicate that both particle size and density should be

as large as possible. In a best-case scenario, the powder would also have the necessary compressibility (indicated by the differences in bulk and tap density) to enable a direct compression approach9. The concern for product performance is that as SDDs are mostly developed with BCS class II APIs (good permeability, poor solubility), the drug product bioavailability challenge is more related to improving solubility. Solubility enhancement may be achieved by increasing the maximum API concentration in solution (BSC class IIb) – here the polymer used in an SDD may play an important role as it may promote supersaturation –

Figure 3: Secondary drying strategy for an SDD (green points: SDD Tg; red line: drying temperature).

6

and/or by improving the API dissolution kinetics (BCS class IIa). Where dissolution kinetics are key to achieving bioavailability, the SDD properties, namely particle size, can play an important role in the dissolution profile of the drug product, in the same way as tablet disintegration due to the erosion of the small SDD granules. Particle size dictates the specific area available for mass transfer of the API in the dissolution medium (the smaller the particle size, the larger the specific area). Bioavailability can also be limited by the supersaturation potential of the amorphous solid dispersion rather than the dissolution kinetics, and in those cases particle size of the SDD is not as critical. Hence, a tight control over particle size should be on the list of priorities of every spray drying scientist when developing an SDD. In the scale-up process from the laboratory to a commercial-scale spray dryer, distinct points should be considered. In the laboratory scale, the bottleneck is usually related to the powder properties that can be attained. Due to the reduced size of the drying chamber, laboratory-scale units typically use external mixing two-fluid nozzles. Such atomisation tends to produce particles with a Dv50 (mean particle size in volume) below 10µm and a bulk density below 0.15g/mL, which are not adequate for downstream processing, primarily due to very

european INDUSTRIAL PHARMACY December 2013

• Issue 19


SCALE-UP OF SPRAY DRIED AMORPHOUS SOLID DISPERSIONS Commercial scale

Optimised laboratory scale

Figure 4: Scanning electron microscopy images comparing powders produced in a commercial scale pharmaceutical spray dryer and an optimised laboratory scale unit with an ultrasonic nozzle.

poor flow properties. Although a certain degree of flexibility exists in the blending step, and despite a judicious selection of the excipients and their relative amounts, it is very unlikely, particularly for large-dose drug products, that a good formulation candidate can be developed without an additional granulation step. In larger-scale spray dryers, an enhanced flexibility exists to improve the SDD properties since the range of atomisation mechanisms is broad, allowing the production of a large spectrum of droplet sizes and consequently products with particle size distributions more suited for downstream processing. During preclinical and formulation screening, with processes being developed on a smaller scale and clinical trials batches being produced with limited powder properties (e.g. small particle size and low bulk density), the advancing of the drug candidates to late-phase stages can pose significant challenges for the spray drying process development. One of these situations may be when an SDD process is locked with a small particle size. An ideal approach would be to have the exact same powder properties across scales, targeting those to be used in late-phase and commercial stages. One such approach involves the use of a laboratory unit with an ultrasonic nozzle atomisation and optimised drying gas flow rate and drying chamber configuration10. The use of ultrasonic atomisation enables the generation of droplets of similar dimensions to those in larger-scale

spray dryers, which coupled with the correct thermal and residence time conditions can meet the objective of matching the larger-scale powder properties in the laboratory. Figure 4 illustrates the morphology of spray dried materials produced in the laboratory and in a larger-scale spray dryer, where particle size and density could be maintained across scales. Droplet size

Experience shows that the most determinant parameter to adjust SDD particle size in spray drying is droplet size. Droplets can be generated through different atomisation mechanisms. The most widely used atomisation mechanisms in pharmaceutical spray drying applications are pressure nozzles and two-fluid nozzles. For the production of SDDs, pressure nozzles are typically preferred due to their flexibility in adjusting droplet size

continued

(although two-fluid nozzles can be more efficient in this respect), and for enabling the production of powders with relatively larger bulk densities than with two-fluid nozzles. Droplet size can be estimated through the use of correlations available in the literature. These correlations are typically developed for water spraying, and conversion for specific solvents can be accomplished by considering their surface tension and viscosity11. While such equations from the literature can be applied successfully for specific geometries (e.g. as for external mixture two-fluid nozzles which have a simpler geometry), their application for vendor-specific pressure nozzle geometries may be less adequate given the particularities of the different nozzles. For such cases, the development of droplet size empirical correlations is one of the most straightforward approaches, provided that droplet size measurements are available. Figure 5 shows droplet size predictions for three different pressure nozzles for atomisation flow rate, using a correlation developed for one such vendor-specific nozzle geometries. This predicts the impact of changing the nozzle on droplet size for a fixed feed flow rate. One of the state-of-the-art methods for droplet size data generation is the use of Phase Doppler analysers. These, however, have limitations for

Figure 5: Droplet size estimation for three different pressure nozzles as a function of feed flow rate.

european INDUSTRIAL PHARMACY December 2013

• Issue 19

7


SCALE-UP OF SPRAY DRIED AMORPHOUS SOLID DISPERSIONS

Figure 6: Thermodynamic space for a large scale spray dryer. The contour colours represent the temperature of the drying gas at the outlet of the drying chamber, while the contour lines are the simulated SDD Tg.

commercial-scale spray dryers due to the very high density of the spray plume (given the very high feed flow rates) which makes the penetration of the laser and adequate measurements of droplet size difficult. In these cases, extrapolation of available correlations may be the only option to make an estimation of droplet size. Macroscopic heat and mass balance

In a macroscopic evaluation of a spray drying process, typically a combined heat and mass balance needs to be considered. In a spray drying process, a solution is fed to the top of a drying chamber where it is atomised into small droplets, being heated and vaporised upon contact with hot drying nitrogen. Solid particles are formed very rapidly, typically within seconds, and then separated from the gas in a cyclone and filter bag; the solvent within is later removed in a condenser, closing the mass balance in terms of solvents and solids. For heat balance closure, the only unknown is the heat transfer coefficient of the drying chamber walls that dictates the amount of heat lost to the surrounding room. As the walls are thermally insulated, the global heat transfer coefficient for the walls is a combination of that of stainless steel and of the

8

insulation material. As such, the global wall heat transfer coefficient for a spray dryer is not easily estimated and needs to be established using actual experimental data in a macroscopic heat and mass balance model. The use of such macroscopic models enables an accurate estimation of the drying conditions that are to be expected (e.g. in terms of relative saturation of the solvent in the drying gas), which, coupled with the data generated in the laboratory in Figure 1, can provide an estimate of the SDD Tg in larger-scale spray dryers as illustrated in Figure 6. Final remarks

The future strategies for the scalingup of SDDs should include the use of advanced computational methods such as CFD that can better anticipate spray-related challenges, e.g. the sticking of materials with low Tg to the walls of the spray dryer. As for the impact of SDD powder properties downstream, key attributes that can affect powder flow (and consequently its processing by blending, roller compaction and tableting) and the SDD dissolution performance need to be better understood so that the spray drying process can be optimised to produce material that meets all downstream requirements. The use of advanced analytical

continued

equipment, such as powder rheometers and compaction simulation apparatuses, should assist in meeting such objectives through the establishment of quantitative relationships between SDD properties (even with small quantities of powder) and downstream performance. Clearly, the possibility of producing SDD in the laboratory with comparable properties to that of larger-scale spray dryers can help to simplify and streamline the process development workflow (from the spray dried powder to the final tablet dissolution performance). The use of special atomisation mechanisms in laboratory-scale spray dryers (such as ultrasonic nozzles) needs to be further investigated to enable an improved match of particle size and density across scales, thereby reducing the time for an SDD formulation to reach the market. References 1

Dobry DE, et al. A model-based methodology for spray-drying process development. J Pharm Innov 2009;4:133–142. 2 Vehring R. Pharmaceutical particle engineering via spray drying. Pharm Res 2008;25:999–1022. 3 Paudel A, et al. Manufacturing of solid dispersions of poorly water soluble drugs by spray drying: formulation and process considerations. Int J Pharm 2013;453:253–284. 4 Ullum T, et al. Predicting spray dryer deposits by CFD and an empirical drying model. Drying Technol 2010;28:723–729. 5 Fletcher DF, et al. What is important in the simulation of spray dryer performance and how do current CFD models perform? Appl Math Model 2006;30:1281–1292. 6 Santos J, Temtem M. Fluid Motion. Pharm Manuf Pack Sourcer 2013; May:74–79. 7 Rong L. Water insoluble drug formulation. London: Taylor & Francis; 2008. 8 Kalogeras I. A novel approach for analyzing glass-transition temperature vs. composition patterns: application to pharmaceutical compound + polymer systems. Eur J Pharm Sci 2011;42:470–483. 9 Qiu Y, et al. Developing solid oral dosage forms. Amsterdam: Elsevier Inc.; 2009. 10 Vanwesenbeeck J, et al. Spray drying scale-up from mg to kg scale with a hydroxypropyl methylcellulose acetate succinate (HPMC-AS) solution. AAPS 2010, poster W5491. 11 Perry RH, Green DW. Perry's chemical engineers' handbook. New York: McGrawHill; 2008.

european INDUSTRIAL PHARMACY December 2013

• Issue 19


THE INNOVATIVE MEDICINES INITIATIVE OrBiTo PROJECT – DEVELOPING THE NEXT GENERATION OF PREDICTIVE TOOLS FOR ORAL BIOPHARMACEUTICS by Mark McAllister, Bertil Abrahamsson, Hans Lennernäs

T

he Innovative Medicines Initiative (IMI) OrBiTo project is a pre-competitive collaboration between pharma, academia and specialist technology companies which aims to enhance our understanding of how orallyadministered drugs are absorbed from the gastrointestinal (GI) tract and apply this knowledge to develop new in vitro tests and in silico models that will better predict the performance of oral formulations in patients. Dr Mark McAllister is a research fellow in the Drug Product Design Group, Pharmaceutical Sciences, Pfizer, located in Sandwich, UK. Mark currently leads the UK Academy of Pharmaceutical Scientists Biopharmaceutics focus group and is Deputy Scientific Coordinator for the Innovative Medicines Initiative ‘OrBiTo’ project.

Introduction

The IMI is a new model for public–private collaboration which aims to support open innovation in pre-competitive research and accelerate the development of safer and more effective medicines for patients. With a €2 billion budget, IMI is the world’s largest public–private partnership in health research and development. IMI is a joint undertaking between the European Union (EU) and the European Federation of Pharmaceutical Industries and Associations (EFPIA), who each contribute €1 billion to IMI in cash (EU) and by in-kind contribution (EFPIA; Figure 1). From the first three IMI calls for projects (spanning 2008–2010), 23 projects were approved for funding involving 221 R&D teams from EFPIA companies, 298 academic institutions, 47 small- and medium-

sized enterprises (SMEs), 11 patient organisations and 7 regulatory agencies. The OrBiTo project was submitted as part of the IMI 4th call for projects in 2011 and, following approval of funding, started in

October 2012 and will run for a 5year period with an overall budget of just over €24 million. The OrBiTo project and consortium

Our understanding of the GI environment and prediction of formulation performance has advanced significantly over the last 15–20 years with the implementation of tools such as in vitro permeability models, biorelevant dissolution media, the biopharmaceutical classification scheme and in silico physiologybased pharmacokinetic (PBPK) models for the prediction of GI drug absorption. However, significant gaps in our understanding of oral biopharmaceutics still limit our ability to select drug molecules and/or formulation approaches which are truly optimised for the oral delivery route1. The impact on preclinical and clinical drug development is often manifested in delays at key decision points due to the need to repeat in vivo trials to confirm formulation or drug product performance. The OrBiTo vision is to transform our ability to predict the in vivo performance of oral drug products across all stages of drug development. This will happen through partnership, collaboration and data sharing, developing our fundamental knowledge of the GI conditions to deliver innovative biopharmaceutics tools which will accurately predict product performance over a range of

Figure 1: Overview of the IMI.

european INDUSTRIAL PHARMACY December 2013

• Issue 19

9


THE INNOVATIVE MEDICINES INITIATIVE OrBiTo PROJECT

continued

Figure 2: Overview of the OrBiTo work packages.

clinically relevant conditions. The integration of in vitro and in silico approaches will provide a biopharmaceutics toolkit, validated using clinical data, to accelerate drug development. The main objectives of the OrBiTo project are as follows. • To increase the understanding of the GI drug absorption process as a prerequisite for improved biopharmaceutical predictions. • To create new or refined in vitro and in silico methods contributing to improved in vivo predictions. • To develop a framework for the optimal use of predictive tools and preclinical models. OrBiTo is structured in four research work packages (WPs) focusing on physico-chemical tools (WP1) 2, in vitro tools (WP2) 3 and in vivo understanding and tools (WP3), with the integration of results and data across all WPs being achieved through the application of in silico models of drug absorption in WP4 (Figure 2) 4. WP1 seeks to improve the understanding of the physicochemical and biopharmaceutical properties, which affect the in vivo performance of poorly soluble active pharmaceutical ingredients (APIs; BCS class II and IV drugs). The data generated by the new

10

models and characterisation tests developed though WP1 will inform the selection of candidate molecules during preclinical testing and guide appropriate formulation selection for first use in human studies. Characterisation data describing biopharmaceutical

properties will also be used as a key input for the integrated in silico models being developed as part of WP4. WP2 will focus on in vitro development tools in the dissolution and permeability area that provide high quality quantitative predictions for late stage/market formulation development. WP3 will focus on generating in vivo understanding in areas where remaining gaps hamper the development of predictive methods. This will include regional permeability studies, enhanced system characterisation of the GI physiological conditions as well as studies of model drugs to elucidate specific biopharmaceutical processes, providing a basis for improved in vitro and in silico tools. In addition to integrating the results from WP 1–3 within PBPK models of the GI absorption process, WP 4 will also seek to build on and improve existing prediction algorithms to improve the accuracy of simulation and

Table 1: OrBiTo project consortium EFPIA member companies AstraZeneca AB, Sweden Abbvie GmbH & CoKG, Germany Bayer Pharma AG, Germany Boehringer Ingelheim International GmbH, Germany GlaxoSmithKline Research and Development Ltd, UK H. Lundbeck A/S, Denmark Janssen Pharmaceutica NV, Belgium Merck Sharp & Dohme Corporation Novartis Pharma AG, Switzerland Orion Corporation, Finland Pfizer Ltd, UK Sanofi-Aventis Research and Development, France

Universities, research organisations, public bodies, non-profit groups Uppsala Universitet, Uppsala, Sweden Copenhagen University, Denmark Ernst Moritz Arndt University Greifswald, Germany Johann Wolfgang GoetheUniversität Frankfurt am Main, Germany Johannes Gutenberg Universität Mainz, Germany Katholieke Universiteit Leuven, Belgium Medical Products Agency, Uppsala, Sweden National and Kapodistrian University of Athens, Greece Netherlands Organization for Applied Scientific Research TNO University of Manchester, UK

SMEs Simcyp Limited, Sheffield, UK Sirius Analytical Ltd, Forest Row, UK Simulations Plus, Inc., Lancaster, USA

University of Strathclyde, UK

european INDUSTRIAL PHARMACY December 2013

• Issue 19


THE INNOVATIVE MEDICINES INITIATIVE OrBiTo PROJECT

prediction of in vivo performance. Much of the work described in WP 1–4 will be underpinned by the development of a novel and unique database containing in vivo data of well-characterised novel APIs and diverse oral pharmaceutical products. It will mainly consist of novel material from the EFPIA partners and the first version of this database is expected to include data from more than 100 clinical studies. EFPIA partners will also supply APIs or formulated drug product for additional experimental characterisation. An important aspect of the research concept is to use this material and database resource across the different WPs in the project to generate common data which can be integrated throughout the programme to identify a generally applicable framework for use of predictive tools. The database will be supplemented by literature data and it will also be open for additions with novel EFPIA compounds during the project. However, it is emphasised that the vast majority of work will be done on novel APIs with the exception of the prospective human clinical studies to be performed within

continued

OrBiTo which will use wellcharacterised marketed APIs as model drugs. The OrBito Project consortium comprises 12 EFPIA companies and an academic–SME group with 14 members as shown in Table 1. Conclusions

Over the 5-year duration of the project, it is hoped that the combination of industrial biopharmaceutics expertise, specialist technology providers and academic centres of excellence will create the next generation of tools for oral biopharmaceutics and transform our ability to predict oral formulation performance. The IMI principles and framework for precompetitive collaboration are central to OrBiTo’s research strategy. This is clearly illustrated by the development of a crosscompany physicochemical and pharmacokinetic database containing extensive and diverse data-sets which will be used to develop and validate new in vitro and in silico tools. In conclusion, the OrBiTo project provides a tremendous opportunity to advance our understanding of oral biopharmaceutics and offers a

template for collaborative efforts in other areas of pharmaceutical science. References 1.Lennernäs H et al. Oral biopharmaceutics tools – time for a new initiative – an introduction to the IMI project OrBiTo. Eur J Pharmaceut Sci 2013, in press (http://dx.doi.org/10.1016/j.ejps.2013.10. 012). 2.Bergström CAS et al. Early pharmaceutical profiling to predict oral drug absorption: current status and unmet needs. Eur J Pharmaceut Sci 2013, in press (http://dx.doi.org/10.1016/j.ejps.2013.10. 015). 3.Kostewicz ES et al. In vitro models for the prediction of in vivo performance of oral dosage forms. Eur J Pharmaceut Sci 2013, in press (http://dx.doi.org/10.1016/j.ejps.2013.08. 024). 4.Kostewicz ES et al. PBPK models for the prediction of in vivo performance of oral dosage forms. Eur J Pharmaceut Sci 2013, in press (http://dx.doi.org/10.1016/j.ejps.2013.09. 008).

Acknowledgements This information is provided on behalf of the IMI OrBiTo consortium.

European Journal of Parenteral & Pharmaceutical Sciences is the official quarterly journal of the Pharmaceutical Healthcare Sciences Society (PHSS). The journal provides a European forum for publishing original papers, editorials and reviews on subjects that cover all aspects of the parenteral and pharmaceutical sciences, both practical and scientific, including:

over 25 over 25 years ye ears of of a advancing dvanc cing pharmaceutical p pharmac harmac ceutical eutic cal and and healthcare h ealthc e ca are a re sciences sscience es

European Journal of Parenteral & Pharmaceutical Sciences 2012 Volume 17 Number 2

■ ■ ■ ■ ■ ■

sterilisation techniques validation microbial detection methods lyophilisation biotechnology LAL testing

■ ■ ■ ■ ■ ■

isolator technology aseptic processing packaging cleanroom design preservatives process filtration

The European Journal of Parenteral & Pharmaceutical Sciences also contains accounts of the scientific meetings, symposia and workshops under the auspices of the PHSS.

Order online at www.euromedcommunications.com Or email: publisher@euromedcommunications.com Tel: +44 (0)1428 752222 Fax: +44 (0)01428 752223

european INDUSTRIAL PHARMACY December 2013

• Issue 19

11


FORMULATION TECHNOLOGY ENABLES THE DELIVERY OF HIV MEDICINES by Peter Timmins, Jonathan Brown

T

he most recent entrants into clinical practice for the treatment of human immunodeficiency virus (HIV) infection include a number of compounds with drug delivery challenges. Innovative formulation technologies have been developed and applied to overcome these challenges and provide medicines suitable for the clinical need. Problems of solubility, pharmacokinetics and physical material properties are described along with their resolution in a series of case examples that are illustrative not only of HIV medicines but are applicable solutions to similar problems in other therapeutic classes. Peter Timmins is head of Portfolio Enabling Technology, a group that is part of Drug Product Science and Technology within the Pharmaceutical Development organisation of Bristol-Myers Squibb Research and Development. He has research interests across the oral drug delivery arena and his group is responsible for developing drug delivery solutions for the small molecule candidates for oral administration that require resolution of solubility or/and pharmacokinetic challenges to enable them for clinical evaluation. Peter’s group has staff and operations in UK and USA. Email: peter.timmins@bms.com Jonathan Brown is a Principal Scientist within the Portfolio Enabling Technology group. He has research interests in oral modified release formulation technologies and physiologically-based pharmacokinetic modelling.

Introduction

Significant advances in the treatment of HIV infection have occurred since its identification around three decades ago, leading to a reduction in its incidence and longer life expectancy of infected individuals1. Although a cure still remains an aspiration, anti-retroviral drugs have been an important factor in managing the infection, and the need for new therapies and improved versions of existing drugs remains important to assure effective viral suppression, to enable patient compliance with medication regimens and to deal with the development of drug resistance1,2. The efforts of drug discovery scientists have generated a number of new anti-HIV agents3 but, as is the case for many emerging drug

12

candidates across all therapeutic areas, the physicochemical and pharmacokinetic properties of a number of the new entities have challenged formulation scientists to assure that the finished medicine delivers the desired clinical profile to realise therapeutic success. This challenge may be further emphasised if doses in the hundreds of milligrams are required, as is the case for a number of HIV therapies. With the emergence of the highly active antiretroviral therapy (HAART) approach in the late 1990s, as newer agents that targeted different stages of viral replication life cycle4 appeared and made this possible, combination drug therapy advanced and eventually provided for simplified treatment regimens with coformulated drugs5.

Assuring the performance of the newer drugs with their inherent challenges, as well as supporting simplified dosing of multiple medicines therefore defines an important role for pharmaceutical formulation. This review focuses on how formulation science has effectively enabled the utility of some newer, as well as some more established, treatments for HIV infection and illustrates examples that inform how to deal with formulation challenges of today’s medicines irrespective of therapeutic class. Formulation of HIV medicines – case histories Efavirenz

Efavirenz (Sustiva, Bristol-Myers Squibb, and a component of Atripla, Bristol-Myers Squibb/Gilead) is a poorly soluble, non-nucleoside reverse transcriptase inhibitor (NNRTI) reported as showing sensitivity of bioavailability to tablet disintegration time. Therefore, the original single entity formulations6 employed optimised disintegrant choices and levels, along with a careful distribution of disintegrant between intragranular and extragranular phases of the tablet, to assure very rapid disintegration. Tablets formulated in this way were bioequivalent to earlier marketed capsule formulations (200mg efavirenz per capsule) and as they contained a higher dose of drug per unit dose form (600mg) in an acceptably sized tablet, offer greater convenience to patients where a dose of 600mg is prescribed. Other work suggested that the visually observed tablet disintegration alone is not the key factor that determines bioavailability for efavirenz, but what is most important is the completion of disintegration down to primary particles and dispersion of the active ingredient7. For two tablet formulations, in vitro disintegration and the time to complete disintegration in vivo as measured by gamma scintigraphy were different, but the one that exhibited slower disintegration both

european INDUSTRIAL PHARMACY December 2013

• Issue 19


FORMULATION TECHNOLOGY ENABLES THE DELIVERY OF HIV MEDICINES

in vitro and in vivo had slower in vitro dissolution but had the highest bioavailability, with higher area under the plasma concentration–time curve, higher maximum plasma concentration and an earlier time to maximum plasma concentration. It was suggested that the reason for the delay in absorption for the apparently faster disintegrating formulation was that it was disintegrating to larger aggregates in vivo relative to the better performing formulation but this difference is not discernible in visual observation of disintegration. From these larger particles, resulting perhaps from poorer effectiveness in this specific formulation of the included sodium starch glycolate disintegrant, drug release was slower. Also it was suggested that in vivo gelation of the sodium starch glycolate acted as a further barrier for drug release7. Translating this learning to a fixed combination dosage form, containing efavirenz with emtricitabine and tenofovir disoproxil fumarate, led to the choice of a bilayer tablet to mitigate compatibility problems and to assure the efavirenz layer could disintegrate readily and disperse the poorly wetting efavirenz particles in vivo to assure bioavailability8,9. A non-bilayer formulation based on wet granulation led to instability, attributed to high water levels

needed for efavirenz granulation that allowed a eutectic formation between emtricitabine and tenofovir disoproxil fumarate which dried as a glassy or amorphous unstable form10. Additionally, interaction with the surfactant included to help wet and disperse the efavirenz was implicated, but its removal led to impaired bioavailability8. A bilayer tablet that separated the efavirenz component showed good stability and bioequivalence of its components to the single entity formulations9. It combines three medicines useful together to treat HIV infection, as they come from more than one therapeutic target class, that have pharmacokinetics suited to once daily dosing and all have the same lack of restriction regarding dosing with food. As such, formulation science has provided for a fixed combination, one tablet, once daily dosing treatment for HIV infection, offering an opportunity to improve patient adherence to therapy which is critical to maintaining disease control11. Etravirine

Etravirine (Intelence, Jannsen-Cilag), a second generation NNRTI, is a very insoluble weak base. Conventional and nanosized drug formulation approaches resulted in negligible blood levels on oral dosing. It was necessary to dose amorphous drug, where the barrier

continued

of crystal lattice breakdown as a first step in dissolution was absent, in order to achieve an acceptable degree of absorption12. However, preparing amorphous drug required cryomilling of crystalline drug for 3 hours at –196°C and the amorphous drug had poor physical stability, being prone to partial recrystallisation13. Due to its low solubility and its low to moderate permeability (Biopharmaceutics Classification System Class IV compound)12, effective drug delivery requires consideration of not only optimising the etravirine physical form, but also to assure very rapid disintegration and in vivo drug release from the dosage form. This is to minimise any impact of variability in gastrointestinal transit time on presentation of the poorly permeable drug to the sites where drug has highest absorption potential (the very high surface area proximal small intestine). An initial formulation for clinical trials that attempted to resolve these drug delivery problems, referred to as a granulo-layered formulation14, required a dose of drug of 800mg twice daily to achieve desired pharmacokinetics. Layering amorphous drug as a dispersion in hydroxypropylmethylcellulose (HPMC) from organic solvent solution onto excipient beads15 was used to improve etravirine dissolution rate, a technology

Figure 1: Process train for manufacture of etravirine tablets based on spray-dried amorphous dispersion from description in innovator patent application18.

european INDUSTRIAL PHARMACY December 2013

• Issue 19

13


FORMULATION TECHNOLOGY ENABLES THE DELIVERY OF HIV MEDICINES

approach previously applied to commercial formulations of the antifungal itraconazole16. This formulation strategy is one of the oral drug delivery approaches described for etravirine in a drug synthesis patent17. Spray drying drug from organic solution together with a polymer was subsequently pursued as an approach to further improve drug dissolution and hence bioavailability, but initial dispersions of drug in HPMC yielded particles on spray drying that were of low bulk density and gave problems with tableting. Co-spray drying etravirine, HPMC and microcrystalline cellulose, with drug and polymer in solution and with microcrystalline cellulose suspended in the spray drying feed, led to denser particles highly suited to processing into tablets (Figure 1)18. This formulation technology provided for significantly improved bioavailability of etravirine in HIVinfected subjects, as indicated by area under the plasma concentration–time curve over the

period to 12 hours post-dosing, and allowed for smaller and a reduced number of tablets to achieve the clinically effective dose. The range of exposure seen with 200mg of the spray dried formulation given twice daily was comparable to that yielded by 800mg of the granulolayered formulation dosed twice daily and with reduced interpatient variability19. Novel HIV attachment inhibitors

Novel HIV attachment inhibitors interfering with the binding of the viral gp120 glycoprotein to the host cell CD4 have been identified by Bristol-Myers Squibb20. One such compound, BMS-626529, is a potent inhibitor of viral replication in vitro but exhibits low oral bioavailability and a very short apparent half-life in vivo. To overcome the low bioavailability of the compound associated with its poor water solubility, a prodrug strategy was pursued in which the chemical structure was modified by the addition of hydrophilic cleavable substituent groups to

continued

improve solubility. Enzymatic hydrolysis in vivo enabled effective delivery of the parent molecule. Based upon animal and human in vivo screening experiments, BMS663068, a phosphonoxymethyl prodrug of BMS-626529, was selected for evaluation in the clinic21. The highly soluble prodrug, when delivered orally, is converted locally via intestinal brush border alkaline phosphatase to the poorly soluble parent, BMS-626529, immediately prior to/during absorption. Maintaining a threshold minimum plasma concentration (Cmin) prior to administration of the next dose is considered important to efficacy, whilst the minimisation of maximum plasma concentration (Cmax) may mitigate peak-related adverse events. Sustained absorption of parent throughout the gastrointestinal tract via slow release of prodrug from a formulation is required to achieve this and overcome the short half-life of the compound. The achievement of target

Figure 2: HME process to make amorphous dispersion formulations (e.g. lopinavir with ritonavir). The HME approach involves dispersing the drug within a pharmaceutically-acceptable polymer in an extruder with the application of heat. If it is done by raising the drug and the polymer in admixture to above the melting point of the drug and the glass transition temperature of the polymer and then cooling without allowing the drug to recrystallise, then the resultant solid dispersion is amorphous. The physical stability of such a formulation might be dependent on the miscibility of the drug and polymer, i.e. the amount of drug dispersed in the polymer, temperature and humidity at which the solid dispersion is stored, and whether there are molecular level interactions between drug and polymer that impair its propensity to recrystallise. Commercial equipment is usually a twin screw extruder which enables feeding of drug and polymer (and other excipients if needed) into the extruder where specifically-designed motor driven screws convey the materials along the heated barrel, mix them, allow them to melt and soften as appropriate, knead and densify them, shear and cut the formed mass and allow it to cool as it leaves the extruder barrel. The resultant material can be further processed into granules and, if needed, blended with other excipients to make it suitable for tableting.

14

european INDUSTRIAL PHARMACY December 2013

• Issue 19


FORMULATION TECHNOLOGY ENABLES THE DELIVERY OF HIV MEDICINES

pharmacokinetics was enabled through the design of a novel extended-release (ER) tablet22. Tablet development was based on in silico modelling and built upon careful evaluation of bioavailability in specific areas of the intestine and colon to gain an understanding of how gastrointestinal physiology and lumenal fluid volume and composition could influence prodrug conversion and absorption of the parent23. The formulation was developed as a non-disintegrating monolithic, matrix tablet utilising hydrophilic swellable cellulose ether polymer technology. Although unbound alkaline phosphatase was shown to penetrate the hydrating gel layer of the tablet and represent an in vivo risk from lumenal alkaline phosphatase, conversion of prodrug to parent within the hydrated dosage form was shown to be inhibited24. This is essential to maintaining the required release rate and pharmacokinetics as premature conversion to parent within the dosage form would lead to reduced bioavailability. The optimised, in vivo stabilised ER formulation was found to be suitable for once to twice daily dosing. Lopinavir with ritonavir

Lopinavir with ritonavir (Kaletra, AbbVie) commercial formulation employs hot melt extrusion (HME), resulting in an amorphous drug dispersion in the finished product. Amorphous dispersions have been progressed as a solution to overcome poor drug-like properties, solubility and dissolution rate, and mitigate poor bioavailability relating to those properties25. HME represents a commercialisable approach to manufacturing such dispersions26 (Figure 2). It has long been used in other industries where a thermoplastic material is softened under temperature and shear force, usually admixed with other materials (fillers, pigments, stabilisers, etc), then extruded to form granules/pellets, tubes, rods or sheets. HME was first described for creating pharmaceutical dosage

forms over 40 years ago27 but only became of demonstrated potential for creation of commercialisable products in the last 15–20 years28. The commercial lopinavir 200mg with ritonavir 50mg combination tablet employs copovidone and sorbitan laurate as the polymeric components to provide the material for tableting. The amount of copovidone in the product was higher than previously used according to the US FDA Inactive Ingredient Listing and required the submission of a package of toxicity studies to support the higher level, including data on repeat dose toxicity, genotoxicity and carcinogenicity29. As well as reducing the number of dosage forms the patient has to take in transferring to the HME formulation from the prior available liquid-filled capsule formulation, the HME formulation avoids the requirement to store the medicine in a refrigerator. The tablet shows lower variability in pharmacokinetic parameters relative to the capsule and has a much diminished food effect, allowing for the possibility of not restricting to dosing with food30,31. These are valuable improvements in terms of supporting patient compliance with this medicine resulting from the application of modern formulation science. Cobicistat

Cobicistat (Tybost, and component of Stribild, Gilead) is a novel cytochrome P450 (CYP) enzyme (CYP3A4) inhibitor used as a “booster” in combination with some HIV treatments, e.g. protease inhibitors, to reduce their metabolism during absorption and so increase the amount of unchanged drug reaching the systemic circulation. Ritonavir is also used as such a boosting agent but cobicistat differs in that it has no anti-viral activity of its own and is purely used to modify pharmacokinetics. Cobicistat drug substance does not occur in crystalline form and is isolated as an amorphous,

european INDUSTRIAL PHARMACY December 2013

• Issue 19

continued

hygroscopic solid foam of low glass transition temperature which readily transforms under ambient conditions via a moisture- and temperature-driven phase transformation into a rubber-like material that is difficult to process into dosage forms. The removal of the absorbed moisture and reversion to the original solid form does not occur32. Initial approaches to making solid dosage forms involved handling the drug substance in solution in an appropriate organic solvent, such as ethanol33. This solution is added to silica and then excipients and water in a high shear mixer to wet granulate. Fluid bed drying achieves removal of the organic solvent as well as the water. The challenges of handling large volumes of flammable solvent in a drug product processing facility, whilst not insurmountable, can be avoided if the drug is loaded on to a suitable carrier material that impart superior handling properties to the drug substance prior to introduction into a dosage form manufacturing area. By adsorbing onto silica by evaporation from a dichloromethane solution of drug as part of the isolation of the cobicistat33, a freeflowing powder is produced. Although still hygroscopic, moisture uptake is now reversible and the cobicistat no longer undergoes phase transformation. The drug adsorbate is highly suited to further processing into pharmaceutical dosage forms. The finished dosage form using the adsorbate showed bioequivalence for cobicistat with dosage forms prepared by the ethanol/water high shear granulation process. For cobicistat, formulation technology has dealt with challenging physical properties of an active pharmaceutical ingredient and provided an approach to improved handling and manufacturing of dosage forms. Nevirapine

Nevirapine, a well-established NNRTI, has recently become available as an ER formulation

15


FORMULATION TECHNOLOGY ENABLES THE DELIVERY OF HIV MEDICINES

(Viramune Prolonged-Release, Boehringer Ingleheim). Although it is inherently a long half-life drug, t½ >45 hours34, there are advantages in offering patients an ER nevirapine formulation. Despite the long halflife, immediate-release nevirapine is typically dosed in treatmentstabilised patients at 200mg twice daily35. A concern with using a 400mg once daily immediaterelease dose is that it results in high mean peak and low mean trough plasma concentrations of drug relative to using a dose of 200mg twice daily, although the exposure to drug as measured by the area under the plasma concentration–time curve was equivalent for once and twice daily dosing regimens. Higher peak concentrations give concern as to an increased risk of liver toxicity and lower trough concentrations raise concern as to risk of emergence of viral resistance35. Also, twice daily dosing is less convenient for HIV patients taking multiple medications where many of their other medicines have once daily dosing. Hence, there is good clinical relevance to creating the ER formulation. Predicting or demonstrating that there is effective drug absorption from the colon is essential to determining the viability of developing an oral ER formulation36. Nevirapine delivered to the ascending and descending colon had 82 and 58% bioavailability, respectively, relative to an orallyadministered suspension of nevirapine37, exceeding the value of ≥40% suggested as an indicator of

likely success38, and supported the development of the ER product. The commercial formulation was selected on the basis of pharmacokinetic evaluation of a series of hydrophilic matrix tablets that employed different amounts and grades of HPMC as a release rate controlling polymer. Tablets contained 20, 25, 30 or 40% w/w of HPMC 2208 4000 cps grade or alternatively 20 or 25% w/w HPMC 2910 4000 cps grade and the amount and grade of polymer used yielded a range of in vitro release rates that translated to a range of in vivo pharmacokinetic profiles39 with the lower amount of polymer leading to a faster release rate, and with the 2910 polymer leading to faster release rates than the 2208 polymer. The marked change in release rate with polymer amount, given similar tablet sizes across the range studied, suggests a degree of dependency on erosion as a key rate controlling mechanism from the hydrated polymer matrix in vitro and in vivo. It was possible to build a correlation between the in vitro release profile and the in vivo performance enabling prediction of the pharmacokinetic behaviour of dosage forms as in vitro release rate was changed, which would enable further formulation development studies to be prosecuted using in vitro drug release rate for decision making40. Contrary to what was seen in the single-dose human study used to establish the in vitro–in vivo correlation, in a steady state human bioavailability study of the 20 and 25% w/w polymer ER formulations,

continued

the (more slowly releasing) 25% w/w polymer formulation showed higher bioavailability compared to the 20% w/w polymer formulation41. As metabolism of nevirapine is impacted by induction of CYP enzymes, which include CYP3A442 (which is more extensively expressed in the upper gastrointestinal tract), a more slowly releasing formulation might deliver more drug to the regions where the enzyme is not expressed compared to the faster releasing formulation, and result in increased exposure that is more obvious in a steady state study. This observation offers the caution that the performance of an enabled formulation has to be considered in light not only of its fabrication technology, but the physiological environment in which it has to operate, including gastrointestinal transit, absorption sites, metabolising enzymes, etc. Summary

The development of modern medicines for the treatment of HIV infection has benefitted from formulation technology to accommodate the inherent properties of the active pharmaceutical ingredient and assure that pharmacokinetic and physiochemical challenges are managed. HIV medicine formulations offer examples of ER technologies, managing in vivo disintegration and dispersion, adsorption onto a carrier, spray drying and HME. All of these technologies are in place for commercialised products and

Clean Air and Containment Review The journal to enhance your knowledge of cleanroom, clean air and containment technology • • • • •

16

Learn about different aspects of these technologies from clearly written articles by experts Keep up to date on standards with regular updates by standards committee members Read about innovations Understand the jargon To subscribe, or for more information including Become an expert yourself contents lists for all previous issues, visit www.cleanairandcontainment.com

european INDUSTRIAL PHARMACY December 2013

• Issue 19


FORMULATION TECHNOLOGY ENABLES THE DELIVERY OF HIV MEDICINES

demonstrate how formulation technology in general may be deployed to deal with the challenges inherent in contemporary potential therapeutic agents of any disease class. References 1

Benzer JA, et al. The current state of HIV therapy. Formulary 2013;48:213–223. 2 Arribas JR, Eron J. Advances in antiretroviral therapy. Curr Opin HIV AIDS 2013;8:1–9. 3 Kumari G, Singh RK. Anti-HIV drug development: structural features and limitations of present day drugs and future challenges in the successful HIV/AIDS treatment. Curr Pharmaceut Design 2013;19:1767–1783. 4 Pomerantz RJ, Horn DL. Twenty years of therapy for HIV-1 infection. Nat Med 2003;9:867–873. 5 Flexner C. HIV drug development: the next 25 years. Nat Rev Drug Discov 2007;6:959–966. 6 Batra U, et al. Compressed tablet formulation. US Patent US7707294; 2006. 7 Gao JZ, et al. Investigation of human pharmacoscintigraphy behaviour of two tablets and a capsule formulation of a high dose, poorly water soluble/highly permeable drug (efavirenz). J Pharm Sci 2007;96:2970–2977. 8 Dahl TC, et al. Unitary pharmaceutical dosage form. US patent application US2007/0099902; 2007. 9 Hussain MA, et al. Development of Atripla®, a fixed dose combination tablet of three drugs: efavirenz, emtricitabine, and tenofovir disoproxil fumarate. Presented at AAPS Annual Meeting, New Orleans, LA, USA; 2010. 10 Dahl TC, et al. Dry granulated composition comprising emtricitabine and tenofovir disoproxil fumarate. European patent specification EP1890681; 2009. 11 Airoldi M, et al. One-pill once-a-day HAART: a simplification strategy that improves adherence and quality of life in HIV-infected patients. Patient Prefer Adherence 2010;4:115–125. 12 Weuts I, et al. Physicochemical properties of the amorphous drug, cast films, and spray dried powders to predict formulation probability of success for solid dispersions: etravirine. J Pharm Sci 2011;100:260–274. 13 Qi S, et al. An investigation into the crystallisation behaviour of an amorphous cryomilled pharmaceutical material above and below the glass transition temperature. J Pharm Sci 2010;99:196–208. 14 Kakuda TN, et al. Assessment of the steady-state pharmacokinetic interaction between etravirine administered as two different formulations and tenofovir disoproxil fumarate in healthy volunteers. HIV Med 2009;10:173–181.

15 Anonymous.

Intelence 100 mg tablets (Tibotec) 01/18/2008 approval [HIV-1]: clinical pharmacology and biopharmaceutics. Sourced from FOI Services, Inc., Gaithersberg, MD, USA; 2008. Downloaded from foiservices.com on April 20, 2010. 16 Gilis PMV, et al. Beads having a core coated with an antifungal and a polymer. US patent 5633015; 1997. 17 De Corte B, et al. HIV replication inhibiting pyrimidines. US patent 7037197; 2006. 18 Kiekens FRI, et al. Process for preparing spray dried formulation of TMC125. US patent application 2009/0197903; 2009. 19 Kakuda TN, et al. Single and multipledose pharmacokinetics of etravirine administered as two different formulations in HIV-1-infected patients. Antiviral Ther 2008;1:655–661. 20 Kadow JF, et al. Discovery and development of HIV-1entry inhibitors that target gp120. In: Antiviral Drugs: From Basic Discovery Through to Clinical Trials (Kazmierski WM, Ed). Hoboken, NJ, USA: Wiley; 2011. 21 Wang T, et al. Use of a phosphonoxymethyl prodrug approach to successfully improve the oral delivery of HIV-1 attachment inhibitors: design, preclinical profile and human exposure. Presented at: American Chemical Society National Meeting, San Francisco CA, USA; 2010. 22 Brown JR, et al. Stable pharmaceutical composition for optimized delivery of an HIV attachment inhibitor. European patent specification EP 2323633; 2012. 23 Brown J, et al. Compartmental absorption modeling and site of absorption studies to determine feasibility of an extendedrelease formulation of an HIV-1 attachment inhibitor phosphate ester prodrug. J Pharm Sci 2013;102:1742–1751. 24 Hanley S, et al. Investigation into the potential for prodrug hydrolysis within a hydrated HPMC matrix. Presented at APS PharmSci Conference, Edinburgh, UK; 2013. 25 Van den Mooter G. The use of amorphous solid dispersions: a formulation strategy to overcome poor solubility and dissolution rate. Drug Discov Today: Technologies 2012;9:e79–e85. 26 Shah S, et al. Melt extrusion with poorly soluble drugs. Int J Pharm 2013;453:233–252. 27 El-Agakey MA, et al. Hot extruded dosage forms. Part 1: technology and dissolution of polymeric matrices. Pharm Acta Helv 1971;46:31–52. 28 Breitenbach J, et al. Solid dispersions by an integrated melt extrusion system. Proc Intl Symp Control Rel Bioactive Mater 1998;25:804–805. 29 EMA. Kaletra, Scientific Discussion, 27 April 2006. Downloaded on 9 October 2013 from www.ema.europa.eu/ema/index.jsp?curl= pages/medicines/human/medicines/00036 8/human_med_000867.jsp&mid=WC0b01 ac058001d124

european INDUSTRIAL PHARMACY December 2013

• Issue 19

continued 30 Breitenbach

J. Melt extrusion can bring new benefits to HIV therapy: the example of Kaletra tablets. Am J Drug Deliv 2006;4:61–64. 31 Klein CE, et al. The tablet formulation of lopinavir/ritonavir provides similar bioavailability to the soft-gelatin capsule formulation with less pharmacokinetic variability and diminished food effect. J Acquir Immune Defic Syndr 2007;44:401–410. 32 EMA. Committee for Medicines for Human Use, Assessment Report: Stribild, 21 March 2013. Downloaded on 18 October 2013 from www.ema.europa.eu/ema/index.jsp?curl= pages/medicines/human/medicines/00257 4/human_med_001654.jsp&mid=WC0b01 ac058001d124 33 Kozlara JM, et al. Use of solid carrier particles to improve the processability of a pharmaceutical agent. US patent application US 2009/0324729; 2009. 34 Lamson MJ, et al. Single dose pharmacokinetics and bioavailability of nevirapine in healthy volunteers. Biopharm Drug Dispos 1999;20:285–291. 35 Cooper CL, van Heeswijk RP. Once-daily nevirapine dosing: a pharmacokinetics, efficacy and safety review. HIV Med 2007;8:1–7. 36 Brown J, et al. Predicting feasibility and characterizing performance of extendedrelease formulations using physiologically based pharmacokinetic modelling. Therapeut Deliv 2012;3:1047–1059. 37 Macha S, et al. Assessment of nevirapine bioavailability from targeted sites in the human gastrointestinal tract. J Clin Pharmacol 2009;49:1417–1425. 38 Connor A, et al. Evaluation of human regional bioavailability to assess whether modified release development is feasible. Presented at AAPS Annual Meeting, San Diego CA, USA; 2007. 39 Cappola ML, et al. Extended release formulation of nevirapine. US patent US 8460704; 2013. 40 Macha S, et al. In vitro–in vivo correlation for nevirapine extended release tablets. Biopharm Drug Dispos 2009;30:542–550. 41 Battegay M, et al. Bioavailability of extended-release nevirapine 400 and 30 mg in HIV-1: a multicenter, open-label study. Clin Ther 2011;33:1308–1320. 42 Lamson M, et al. Nevirapine induces both CYP3A4 and CYP2B6 metabolic pathways. Clin Pharm Ther 1999;65:137.

17


TAKING STEPS TO IMPLEMENT THE FALSIFIED MEDICINES DIRECTIVE AND COMBAT COUNTERFEITING IN EUROPE – THE EUROPEAN STAKEHOLDER MODEL by Richard Bergström

F

alsified medicines present a public health threat at global level, with counterfeit goods endangering the health of patients in countries around the world. With the increased prevalence of unlicensed online “pharmacies” and points-of-sale, the problem is attracting growing attention – inspiring governments, security officials and the pharmaceutical industry to take action. The pharmaceutical industry has a vested interest in fighting counterfeiting: developing new medicines is only one part of providing healthcare treatments for patients – we also have to ensure that people benefit from those medicines. Part of that involves making sure that any medicine we produce reaches the patient in the same condition as when it leaves the manufacturing site. Richard Bergström was appointed as Director General of the EFPIA in April 2011. Over the past 20 years he has worked for Roche, Novartis and with the Swedish pharmaceutical industry association (LIF). Since 2006, he has been an Advisor to the WHO on good governance in medicine.

Falsified Medicines Directive: fighting a growing problem

Counterfeiting is a crime with very real consequences: it presents a major threat to patient safety and we have seen the devastating evidence of this. Just this past October, customs agents seized 1 million fake Xanax pills at Zurich airport1. In May of this year, French customs officers seized 1.2 million doses of counterfeit aspirin – an over-thecounter medicine that could have reached patients even more easily than the fake anti-anxiety pills seized in Switzerland 2. Germany, meanwhile, experienced one of its most significant counterfeiting scares in March, when it was discovered

that a common heartburn counterfeit or sub-standard medication had made it onto the market3. In 2011 alone, approximately 30 million fake medicines were seized at EU borders4. What makes these statistics even more troubling is the fact that it is impossible to know what the real numbers are – but we can be sure they are much higher than what is reported. The people behind counterfeiting are criminals – they aren’t reporting earnings and sales, they aren’t releasing quarterly reports. It is impossible to gauge just how big the problem is. And that is cause for serious concern. All we can know is that we need to act – and quickly.

In an attempt to fight back against counterfeiters and the dangerous goods they bring into the markets, the European Union (EU) put forth the Falsified Medicines Directive (FMD; 2011/62/EU) first published in the EU Official Journal on 1 July 2011. The FMD sets out steps to secure the supply chain of medicines in Europe. It asserts that all prescription-only medicines will have to bear safety features (i.e. a unique serial number placed on each pack, together with tamper-evident packaging)*. The FMD also requires the establishment and management of a repositories system that will store the unique identifiers of the serial packs, and contain information on the safety features. The European Commission will determine the specifications of the serial number to be placed on packs when it sets out the rules for implementation in the “Delegated Acts”. Once the Delegated Acts are published – anticipated in the second half of 2014 – EU Member States and pharmaceutical companies supplying the EU market will have 3 years to take the necessary steps and ensure they are in compliance. A solution in the European Stakeholder Model

In Europe, the FMD is an important step in better protecting patients from counterfeit medicines. The European Federation of Pharmaceutical Industries and Associations (EFPIA) sees the need to move as quickly as possible to ensure the legitimate supply chain is as safe as possible. One of the FMD’s key elements is the onus it places on pharmacists to be able to verify that the medicine they dispense to patients is genuine. Towards this end, EFPIA has joined together with the Pharmaceutical Group of the European Union (PGEU), the Groupement International de la Repartition Pharmaceutique (GIRP) and the European Association of the Euro-Pharmaceutical Companies (EAEPC) to develop the European Stakeholder Model (ESM) Medicines Verification System. These organisations – representing

* Certain products or product categories of prescription-only medicines might be exempted according to a risk assessment. Over-thecounter medicines, for instance, are excluded – unless there is a risk of falsification.

18

european INDUSTRIAL PHARMACY December 2013

• Issue 19


TAKING STEPS TO IMPLEMENT THE FALSIFIED MEDICINES DIRECTIVE

Figure 1: The European Medicines Verification System (EMVS)†.

pharmacists, wholesalers, and parallel traders, respectively – came together with the aim of developing a system that will provide a high level of security for patients while being costeffective, pan-European and interoperable, and capable of being effectively integrated into existing structures and practices in the distribution chain. The result is the European Medicines Verification System (EMVS; Figure 1), a system designed to ensure the medicines are making it safely from the point of manufacture to the point of sale – to the patient. The EMVS proposed by EFPIA, EAEPC, PGEU and GIRP is comprised of the European Hub and the National Blueprint Systems (nBPS). The European central hub is connected to a series of singlecountry or multi-country data repositories that serve as verification platforms; pharmacies and other registered parties can use these to check a product’s authenticity. The system will be interoperable between EU countries and will allow for the reconciliation of products traded between EU member states (known as parallel traded products) through the European central hub. It will also offer those countries that do not want to set up their own national system the opportunity to join an existing product verification infrastructure. These components are to be managed by the European Medicines Verification Organisation

(EMVO), which is to be founded in the course of 2014 and foresees participation of authorities and other relevant stakeholders in the overall governance. The EMVS was successfully tested in a pilot project in Sweden from 2009–2010. Stakeholders in Denmark, Finland, Norway and Sweden have already expressed their commitment to the Blueprint System. Meanwhile, development and implementation of the European Hub is well under way. By the first quarter of 2014, results are expected from user-accepted testing in Germany, in which the Hub was connected to the German securPharm system†, demonstrating the first scenario covering the full information chain at European level – from manufacturer to pharmacy. Fighting counterfeiting in a collaborative and comprehensive way

The measures set forth by the FMD, and the systems being developed to help implement its provisions are a major step forward in the EU against counterfeiting. But fighting counterfeiting requires a collaborative, multi-faceted effort. Part of this simply involves education: when it comes to the dangers of counterfeit medicines sold on the Internet, for instance, patients need to be aware that the vast majority of medicines they come across on the Internet are

continued

either counterfeit or unsafe. The pharmaceutical industry invests huge amounts of time and money into developing medicines to treat a huge variety of illnesses – but this work is irrelevant if we can’t ensure that patients are safely receiving these medicines. Any medicine that reaches the patient should be in the same condition as when it leaves the manufacturing site. That is the goal of initiatives like the ESM and the FMD. The sooner we can implement such measures, the sooner we will be making patients that much safer. References 1

2

3

4

BBC. Fake Xanax anxiety pills from China seized in Zurich; 18 October 2013. www.bbc.co.uk/news/world-europe24585099 EUbusiness. Million doses of fake aspirin from China seized in France; 25 May 2013. www.eubusiness.com/newseu/france-china.or9/ Die Welt. Razzia wegen eines gefälschten Medikaments; 7 March 2013. www.welt.de/newsticker/news3/article114 240342/Razzia-wegen-eines-gefaelschtenMedikaments.html European Commission. Report on EU customs enforcement of intellectual property rights. Results at the EU border – 2011; July 2012. http://ec.europa.eu/taxation_customs/res ources/documents/customs/customs_cont rols/counterfeit_piracy/statistics/2012_ipr_ statistics_en.pdf

Editor’s comment:

EIPG, a stakeholder organisation representing the industrial pharmacists responsible for ensuring, under Art. 51 of the Directive, that the safety features have been affixed to the packaging of medicinal products, commented on the EMVS during a symposium at the University of Lisbon in 2012. In particular, EIPG expressed its concern that the model proposed only voluntary verification at the wholesaler dealer level, given that Art. 80(ca) of the Directive stipulates that "Wholesale distributors must verify that the medicinal products received are not falsified by checking the safety features on the outer packaging”.

† securPharm is a national anti-counterfeiting initiative. Launched by German pharmaceutical manufacturing, pharmacist and wholesaler associations, it is designed to test whether medicinal products are genuine or not and is intended to comply with the EU FMD.

european INDUSTRIAL PHARMACY December 2013

• Issue 19

19


regulatory review The current review period has seen a number of changes in the regulation of medicines and regulatory guidance in the EU, USA and International markets.

USA Proposed regulation – administrative detention authority during inspection of drugs intended for human or animal use

Under this proposed regulation, the Food and Drug Administration (FDA) will be able to administratively detain drugs encountered during an inspection that an officer or employee conducting the inspection has reason to believe are adulterated or misbranded until FDA has had time to consider what action it should take concerning the drugs, and to initiate legal action, if appropriate. Draft Guidance for Industry – Specification of the Unique Facility Identifier (UFI) System for Drug Establishment Registration

FDA’s preferred UFI for a drug establishment is the Data Universal Numbering System (DUNS) number. The DUNS number is available free of charge to all drug establishments. Alternative identifiers may only be used after consultation with FDA. Unique Device Identification (UDI)

FDA has released a final rule requiring that most medical devices and certain combination products that contain devices that are distributed in the USA carry a UDI. Secure Supply Chain Pilot Program (SSCPP)

Qualified firms will be enabled to expedite the importation of active pharmaceutical ingredients and finished drug products into the USA. Participating firms in this voluntary programme must meet certain criteria. The pilot program will run from February 2014 through February 2016.

20

Draft Guidance for Industry – Refuse-to-Receive Standards

This guidance is intended to assist sponsors preparing to submit abbreviated new drug applications (ANDAs) and prior approval supplements to ANDAs for which the applicant is seeking approval of a new strength of the drug product. The guidance describes what should be included and highlights serious deficiencies that may cause refusal to receive an ANDA.

Europe Q&A – Practical implementation guidelines on variations

Procedural elements in relation to the implementation of the revised guidelines are clarified. Improving the safety of medical devices

Following the Poly Implant Prothèse breast implants and other scandals the European Commission has adopted two measures for immediate implementation. The new rules are: • a Commission Implementation Regulation clarifying the criteria to be met by notified bodies; • a Recommendation clarifying the tasks these bodies have to undertake when they perform audits and assessments. Reflection paper – coated nanomedicines general issues for consideration

The reflection paper describes general issues to consider during the development of nanomedicines that have a coating.

MHRA Can a single system simplify reporting of incidents to the MHRA?

The MHRA currently runs five reporting systems to collect reports of different types of incidents involving medicines, medical devices and blood. MHRA is considering whether bringing all

five systems together under the Yellow Card Scheme brand (currently used for reporting side effects to medicines) could help increase incident reporting and reduce confusion of reporters. Proposed introduction of Telland-Do variations for a specific subset of parallel import licenses

Parallel importers (PIs) must notify MHRA if they observe certain changes to the imported products (e.g. changed appearance of dosage forms). While MHRA is investigating the changes, there is a ‘stop-processing’ requirement on the importing companies which prohibits them from processing, repackaging or releasing the product onto the market until given approval. This can be an unexpected disruption of several months and affect the importer’s business negatively. The new proposal abolishes the ‘stop-processing’ requirement if imported products are part of European Mutual Recognition or De-Centralised procedures. PIs will notify the Agency of the changes and release the products onto the UK market. This notification principle is known as ‘Tell-and-Do’. Falsifed Medicines Directive (FMD)

The UK MHRA has published on its website an overview of the medicines regulation and guidance in relation to the FMD as it applies to the UK. Nicotine-containing products

The European parliament did not support The Commission’s proposal to regulate electronic cigarettes as medicines. MHRA, however, will continue to encourage companies voluntarily to seek a licence for their products. Q&A for specials manufacturers

Q&As have been used in this guidance to promote easy updates Continued on page 21

european INDUSTRIAL PHARMACY December 2013

• Issue 19


news from the EIPG US Pharmacopoeial Convention

EIPG has received a formal invitation to be an observer organisation at the US Pharmacopoeial Convention. The organisation is looking for European candidates for the USP Council of Experts and Expert Committees to serve for the period of 2015–2020. Anyone wanting to impact public health, share expertise and collaborate with colleagues worldwide and add to their career experiences can apply online during the coming year (www.usp.org/) Please let us know if you are a member of your National Association within EIPG and are applying to become an expert for the USP.

Medicines Agency’s (EMA’s) dedicated facilities workshop attended by Piero Iamartino (VP, Italy) was discussed. European Commission

Representatives of EIPG and the community and hospital pharmacists (PGEU and EAHP) attended a meeting with Dr Patrizia Tosetti, DG SANCO, to discuss medicines shortages. Various EIPG concerns were raised and some suggestions were made to her. EMA a. Comments on the revision of the GMP guidelines

The President and Treasurer’s visit to Bulgaria was reported and practical aspects of the General Assembly 2014 were considered. A series of points on medicines shortages were drafted including the working of Article 126a. The European

EIPG submitted comments on the revision of the GMP guidelines (Annex 16) on certification by a QP and batch release. b. Interested Parties Meeting Claude Farrugia (VP, Malta) attended on behalf of EIPG. The EMA Workplan was discussed and David Cockburn, chair of the meeting, confirmed that cooperation and collaboration with industry is one of their fixed topics for consideration. Presentations were made by representatives from Rx-360 and EXCiPACT on their auditing programmes, supply chain security and certification scheme for active pharmaceutical ingredients (APIs) and excipients. APIC, the APIs Committee presented on their development of a “how to do” document to help industry to comply with good distribution practice (GDP) for APIs. A representative from EFPIA requested clarity on the

Regulatory Review

International

Continued from page 20

WHO

Future training opportunities

Discussions are underway between EIPG and representatives of a training company which covers qualified persons (QP)/quality, quality by design and biopharmaceuticals aimed, in particular, at professionals who are working in companies operating in niche areas. The aim is to offer ondemand training for the members of EIPG Associations and those who visit our EIPG website. October EIPG Bureau Meeting

when further clarification on specific good manufacturing practice topics is required relating to the manufacture of unlicensed medicines. It does not replace any of the requirements already contained in Guidance Note 14.

Good trade and distribution practices for pharmaceutical starting materials (revision)

A number of recent incidents have created awareness of the need for further improvement of the present guidelines. A new draft has been issued to a restricted audience for comment.

european INDUSTRIAL PHARMACY December 2013

• Issue 19

interpretation of several requirements for GDP. Further information will be published on the discussions held during this meeting. c. Dedicated Facilities Workshop A report from Piero Iamartino (VP, Italy) who participated on behalf of EIPG is as follows. The revised versions of EU GMP Chapters 3 and 5, together with the EMA-proposed guidance about a toxicological tool in evaluating dedicated manufacturing facilities, were discussed between EMA, its regulatory partners and the representatives of industry coordinated by EFPIA. The need to set health-based limits and to adopt quality risk management principles were agreed among all participants. However, industrial representatives requested a more flexible approach, though always being adequately justified. Amendments of the text were requested for IMPs (low toxicological data available), APIs (specific guidance to be added), genotoxic materials (threshold limit to be in line with ICH M7). According to industry, a suitable implementation period is necessary for existing products, while the application to “new products” (definition still to be clarified) could start with usual timeframes after the guidance has been finalised. Authorities will provide a revision of the three documents and a second public consultation is expected to be held in 2014. Jane Nicholson, Executive Director EIPG, jane@nicholj.plus.com

For further information on these and other topics we suggest you refer to the websites of relevant regulatory bodies and to current and past editions of “GMP Review News” published by Euromed Communications. To subscribe to this monthly news service contact info@euromedcommunications.com

21


Everything you need to know about the operation and management of cleanrooms Edited by Tim Sandle and Madhu Raju Saghee In 26 chapters and over 600 pages this book provides a unique tool to help you achieve regulatory compliance. It first creates a foundation in history and established practice and then helps you understand how state of the art technology and engineering solutions can deliver the best practice and so provide reliable systems performance.

Wide application

ALS C I T EU E C R A A M HAR THC NP

I

L A E H

AND

Edi

ted

by

Tim

dl San

nd ea

Ma

Raj dhu

ag uS

hee

Covers all aspects of cleanroom operations for pharmaceutical cleanrooms, hospital pharmacies and research laboratories.

aspect of cleanroom management, each providing practical advice and information.

Expert advice

International

The chapters are written by acknowledged international experts from companies involved in every

The book includes guidance to regulatory approaches and legislation in Europe, Asia and the USA

"This book is unlike any other cleanroom-related book I have read, in that it contains all the necessary information in one publication. ... each of the chapters contains an abundance of detailed information on the specific topic, with excellent references." European Journal of Parenteral and Pharmaceutical Sciences

order online at www.euromedcommunications.com Or contact the publishers: email: publisher@euromedcommunications.com; Tel: +44 (0)1428 752222; Fax: +44 (0)1428 752223.


events JANUARY 21-23 January 2014 – Frankfurt, Germany Clinical Supply Chain www.pharma-iq.com 22-24 January 2014 – London, UK Understanding and Preparing the Quality and Pharmaceutical Module www.pti-global.co.uk 28-29 January 2014 – Berlin, Germany Protective Packaging Solutions for Pharmaceutical Product Stability www.gmp-compliance.org 28-29 January 2014 – London, UK Joint Regulators/Industry QbD Workshop www.pda.org 28-29 January 2014 – Washington, DC USA 11th Annual Pharmaceutical Compliance Congress www.cbinet.com

18-20 February 2014 – Munich, Germany 6th Disposable Solutions for Biomanufacturing Summit www.disposablebiomanufacturing. com

24-26 March 2014 – Heidelberg, Germany ICH Q7 Compliance for APIs Manufactured by Chemical Synthesis www.gmp-compliance.org

24-25 February 2014 – Washington DC, USA 2014 Aseptic Annual Conference www.ispe.org

25-26 March 2014 – Lyon, France Modern Biopharmaceutical Manufacturing www.pda.org

24-27 February 2014 – Montreal, Quebec, Canada 12th Annual Cold Chain GDP & Temperature Management Logistics Summit www.coldchainpharm.com

26-27 March 2014 – Frankfurt, Germany 4th Annual Pharma Packaging & Labeling Forum www.flemingeurope.com

25-26 February 2014 – Barnard Castle, UK Cleaning Validation www.honeyman.co.uk 27-28 February 2014 – London, UK Process Validation for Pharmaceutical Products Regulated by EMA & FDA – Balancing Science and Risk during the Product Lifecycle www.management-forum.co.uk

FEBRUARY 4-5 February 2014 – London, UK EU Biosimilar Registration and Marketing Requirements www.pti-global.co.uk 6 February 2014 – London, UK Developments in Analysis of Orally Inhaled and Nasal Drug Products www.jpag.org

MARCH 4-5 March 2014 – London, UK Clinical Outsourcing & Partnership World 2014 www.healthnetworkcommunicatio ns.com 11-12 March – Brussels, Belgium Parenteral Packaging www.pda.org

26-28 March 2014 – Barcelona, Spain 19th Congress of the EAHP: The Innovative Hospital Pharmacist – Imagination, Skills and Organisation www.eahp.eu 31 March - 3 April 2014 – Lisbon, Portugal 9th Pharmaceuticals, Biopharmaceutics and Pharmaceutical Technology World Meeting www.apv-mainz.de

APRIL 1-4 April 2014 – Barcelona, Spain World Generic Medicines Congress Europe 2014 www.healthnetworkcommunicatio ns.com

11 February 2014 – Manchester, UK Good Clinical Practice Symposium www.mhra.gov.uk

13 March 2014 – London, UK The Pharma Summit 2014 – Reinventing Business Models and Markets www.economistconferences.co.uk

1-4 April 2014 – Barcelona, Spain Biosimilar Drug Development World Europe www.healthnetworkcommunicatio ns.com

18-19 February 2014 – Brussels, Belgium 6th Annual Disposable Solutions for Biomanufacturing www.pharma-iq.com

17-18 March 2014 – London, UK Distribution of Medicines – The New EU Good Distribution Practice Guide...Are You Prepared? www.management-forum.co.uk

2-3 April 2014 – Prague, Czech Republic 10th Annual BioProcess International European Summit www.informa-ls.com

18-19 February 2014 – Berlin, Germany Pharmaceutical Microbiology www.pda.org

14 March 2014 – London, UK Good Clinical Practice Symposium 2014 www.mhra.gov.uk

2-3 April 2014 – Barcelona, Spain World Generic Medicines Congress Europe 2014 www.terrapinn.com

european INDUSTRIAL PHARMACY December 2013

• Issue 19

23


Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.