03.05.2015 Views

IPI - Editorial

IPI - Editorial

IPI - Editorial

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>IPI</strong><br />

International Pharmaceutical Industry Spring 2009<br />

The 2008 Revised Declaration of Helsinki<br />

and Proposed Solutions for Research<br />

Ethics in Globalised Clinical Trials<br />

Medical Imaging and<br />

Electronic Data Capture<br />

in Clinical Trials: the<br />

Future Paradigm<br />

Drug Counterfeits<br />

Subject of an Experts<br />

Discussion at the<br />

European Parliament<br />

Managing your info<br />

flow with LIMS<br />

www.ipimedia.com


<strong>IPI</strong><br />

<strong>IPI</strong><br />

International Pharmaceutical Industry<br />

Contents<br />

EDITOR:<br />

Dr Patricia Lobo MSC, PhD<br />

E-mail: patricia.lobo@aol.com<br />

DIRECTORS:<br />

Martin Wright<br />

Mark A Barker<br />

PUBLISHER:<br />

Clive Baigent<br />

EDITORIAL ASSISTANT:<br />

Linda Stewart<br />

E-mail: linda@ipimedia.com<br />

BUSINESS DEVELOPMENT:<br />

Anthony Stewart, Marie Powell<br />

DESIGN DIRECTOR:<br />

Ricky Elizabeth<br />

CIRCULATION MANAGER:<br />

Dorothy Brooks<br />

E-mail: Dorothy@ipimedia.com<br />

FINANCE DEPARTMENT:<br />

Martin@ipimedia.com<br />

RESEARCH & CIRCULATION:<br />

Gramatikov-Vilian@ipimedia.com<br />

COVER PHOTOS: In-House productions<br />

PRINTED BY:<br />

SW TWO, UK<br />

www.swtwo.com<br />

PUBLISHED BY: Pharma publications<br />

www.ipimedia.com<br />

Diamond Key building, Unit 4, Burwell,<br />

industrial estate, Burwell road, London E10 7QG<br />

Tel: 0044 (0)208 591 7584,<br />

Fax: 001 (480)247 5316<br />

E-mail: info@<strong>IPI</strong>media.com<br />

www.<strong>IPI</strong>media.com<br />

All rights reserved. No part of this publication may<br />

be reproduced, duplicated, stored in any retrieval<br />

system or transmitted in any form by any means<br />

without prior written permission of the Publishers.<br />

The next issue of <strong>IPI</strong> will be published in summer<br />

2009 and quarterly thereafter.<br />

issn no International pharmaceutical industry<br />

ISSN 1755-4578<br />

The opinions and views expressed by the authors<br />

in this magazine are not necessarily those of the<br />

Editor or the Publisher. Please note that although<br />

care is taken in preparation of this publication, the<br />

Editor and the Publisher are not responsible for<br />

opinions, views and inaccuracies in the articles.<br />

Great care is taken with regards to artwork<br />

supplied, the publisher cannot be held responsible<br />

for any loss or damage incurred. This publication<br />

is protected by copyright.<br />

2009 PHARMA PUBLICATIONS<br />

6 EDITORS LETTER<br />

REGULATORY AND MARKETPLACE<br />

8 America’s Pharmaceutical Research Companies:<br />

Improving Patient and Practitioner Knowledge about<br />

Prescription Medicines<br />

When it comes to providing patients with excellent healthcare,<br />

there is one essential ingredient that everyone in the healthcare<br />

delivery chain can subscribe to: informed, knowledgeable<br />

patients, active in their own treatment, make the best patients.<br />

Billy Tauzin President and CEO of the Pharmaceutical Research<br />

and Manufacturers of America (PhRMA) Explains - one sure<br />

way to improve the quality and utility of healthcare across the<br />

board is to empower patients with information and help them to<br />

be true partners in their own care.<br />

12 The 2008 Revised Declaration of Helsinki and Proposed<br />

Solutions for Research Ethics in Globalised Clinical Trials<br />

In October 2008 the World Medical Association (WMA) once<br />

again revised the Declaration of Helsinki (DoH), this time at its<br />

General Assembly Meeting in Seoul, South Korea. While the<br />

WMA insisted that this revision would be largely for editorial<br />

purposes, the result is a significantly altered 'Ethical Principles<br />

for Medical Research Involving Human Subjects’, the current<br />

subtitle of the DoH. Francis Crawley of GCP Alliance discusses<br />

the changes and its implications.<br />

16 e-Networking in the Drug Discovery Industry<br />

The complexities of modern drug discovery often require<br />

cooperation between many different participants, sometimes<br />

located in different continents. This is particularly true in an<br />

outsourcing environment, where clinical trials, manufacturing<br />

and even preclinical research become globalised. Demands for<br />

effective communication between companies, laboratories and<br />

individuals have therefore increased enormously. Fortuitously, or<br />

possibly causatively, these demands coincide with the supply of<br />

internet-based networking tools that make geographical<br />

boundaries and time zones irrelevant. Dr. David Bailey and<br />

Edward Zanders of Biovillage Ltd discuss why e-Networking is<br />

expanding rapidly and is here to stay.<br />

DRUG DISCOVERY, DEVELOPMENT & DELIVERY<br />

20 Strategies for the formulation of poorly soluble<br />

compounds and polymorph drugs based on<br />

polyelectrolyte carriers<br />

The main challenge in the administration of poorly soluble<br />

drugs is to design oral formulations displaying a relevant<br />

bioavailability and to develop injectable systems for the<br />

parenteral application with minimized toxicity and high drug<br />

loading. The LBL-Technology ® offers a strategy for enhancing<br />

the kinetic of dissolution of BCS class II drugs based on the<br />

presentation of the drug in the form of nanoparticles stabilized<br />

with layers of polyelectrolyte complexes. Dr. Maria Gonzales -<br />

Ferreiro of Capsulation Pharma explains.<br />

<strong>IPI</strong> 1


<strong>IPI</strong><br />

Contents<br />

CLINICAL RESEARCH<br />

23 Clinical Research Education & training: opportunties and issues<br />

The demand for the clinical research professionals is rising globally<br />

and clinical trials are being outsourced from several countries,<br />

including rapidly growing emerging markets like India and China.<br />

As a result, human resource managers have a challenge in front of<br />

them to find out or develop the right quality of manpower. Kamal<br />

Sahani of Cliniminds explains why in addition to the hiring<br />

experienced professionals, they need to consider clinical research<br />

professionals who have undertaken formal training or education in<br />

clinical research.<br />

26 Patient Recruitment and Retention in Clinical Trials<br />

Patient recruitment and retention is the leading problem drug<br />

companies’ face in developing their new pipelines. As recruiting<br />

becomes more diverse and complex, pharmaceutical companies<br />

are striving to discover new innovative ways to facilitate recruitment<br />

and keep patients enrolled in the clinical trial. With more and more<br />

clinical trials being done on a global level, it is imperative to have<br />

an understanding of both government regulations and social<br />

protocol that accompany these new markets. Dr. Rajam Jaishanker<br />

of Quest Labs analyses how, speeding up clinical trials is one<br />

opportunity to cut costs, improve efficiency and bring new drugs to<br />

market faster.<br />

30 Medical Imaging and Electronic Data Capture in Clinical<br />

Trials: the Future Paradigm<br />

All clinical trial data are ultimately digital. The pathway to this<br />

digital database is not always as connected as one might expect in<br />

this technological age. The backbone of clinical trials is now<br />

Electronic Data Capture (EDC), and this is not yet synchronous with<br />

the other major aspects of data collection, such as medical<br />

imaging. Dr. Collin Miller of Bio Imaging explores the combination<br />

of medical imaging with EDC and provides the future paradigm for<br />

the triallist in the digital age.<br />

34 Providing regulatory submissions in electronic format – US<br />

and EMEA actions<br />

The United States Food and Drug Administration (FDA) is taking<br />

concrete steps to require the submission of all types of regulatory<br />

communication in electronic format. Regulatory authorities globally<br />

are all moving to require electronic submission of regulatory<br />

communication. The reasons are many but the public safety<br />

responsibilities of the regulators require significantly faster access<br />

to content than can be supported by paper format. Antoinette<br />

Azevedo shows how great strides have been made by the<br />

regulatory authorities in the US and EMEA to prepare their<br />

technical infrastructures for eCTD.<br />

2 <strong>IPI</strong><br />

LABS<br />

38 Managing your info flow with LIMS<br />

Providing the backbone of a clinical trials organisation, a<br />

Laboratory Information Management System (LIMS) impacts on all<br />

departments, machinery and personnel. In today's market no<br />

clinical laboratory or CRO could operate without the support of a<br />

well integrated LIMS package. And as the technology gets even<br />

more sophisticated, so do the opportunities for the pharmaceutical<br />

industry. Sam Singh of ACM Pivotal explores the evolution of<br />

Laboratory Information Management Systems (LIMS) from the point<br />

of view of a global clinical trials organisation.


<strong>IPI</strong><br />

Contents<br />

IT LOGISTICS<br />

42 E-freight – Solutions for tough times<br />

The SWISS approach towards saving the environment and manage<br />

cargo consignments in electronic form.<br />

In the complex world of international air freight, where profitability is<br />

measured by minutes saved, kilograms shipped and litres of fuel<br />

burned, the Swiss penchant for precision and reliability is a<br />

valuable asset. Bernd Maresch of Swiss WorldCargo explains why<br />

it has indeed given the airline a steady foundation that has seen<br />

profits grow, even as the industry as a whole continues to tighten<br />

its belt.<br />

48 Risk Management in Clinical Trial Shipping<br />

Every time clinical trial supplies or the samples which are<br />

generated during the lifetime of a trial are transported then an<br />

element of control is taken from your hands. In order to minimise<br />

the lack of control, Sue Lee of World Courier discusses what kind<br />

of risks shipments face when they are actually in transit and when<br />

they get to the site, so that an assessment can be made of those<br />

risks, and contingency planning can be used to reduce them as<br />

much as is feasible in a time- and cost-efficient manner.<br />

4 <strong>IPI</strong><br />

MANUFACTURING & PACKAGING<br />

52 Greater flexibility in pharma packaging by reducing lot sizes<br />

and increasing machine running times<br />

In numerous pharma companies, the following problem is being<br />

posed: shrinking lot sizes are constantly worsening the ratio of<br />

packaging system set-up time to running time. The contribution<br />

below brings into question the current customary 1:1 linking of a<br />

blister machine, inline printing system and cartoning machine and<br />

looks at the feasibility and possibilities of partially decoupled<br />

packaging processes. Dr. Ralph Blum of MediSeal GmbH shows<br />

when packaging small production lots, partially decoupling lines<br />

and linking the individual line segments with a logistics module<br />

leads to a significantly better utilisation of capacity and distinctly<br />

shorter throughput times.<br />

56 Drug Counterfeits – Subject of an Experts Discussion at the<br />

European Parliament.<br />

The European Parliament and chairman of the Life Science Circle<br />

invited experts and stakeholders to discuss the threats of drug<br />

counterfeits in Europe. The venue, which took place at the<br />

European Parliament in Brussels on December 9th 2008,<br />

highlighted the increasing concerns about fake medicines<br />

penetrating the supply chain and the urgency for measures to<br />

prevent a possibly catastrophic loss of confidence in medicines<br />

with all the negative consequences on health conditions of<br />

individuals and on public health. Tassilo Korab of HCPC Europe<br />

analyses the outcome and proposed solutions.<br />

60 Pharmaceutical piracy<br />

Tracking and Tracing and Anti-Counterfeiting<br />

In 2010, counterfeit drug sales are estimated to reach 75 billion US-<br />

Dollars globally, an increase of more than 90 percent from 2005. This<br />

figure is highly alarming. But most alarmingly, we must realise that this<br />

is no long-term prediction anymore. Nicole Golomb of Simons<br />

Security Systems GmbH (3S) explains why, the pharmaceutical<br />

industry, politics, organisations and associations not only need to<br />

get plans going for an effective protection of medical products<br />

– a prompt implementation is also necessary as soon as possible.


<strong>IPI</strong><br />

Editor’s letter<br />

<strong>IPI</strong> – The global forum for Life<br />

Science in 2009 and beyond…<br />

I’m delighted to join the team at<br />

International Pharmaceutical Industry (<strong>IPI</strong>)<br />

published by Pharma Publications<br />

(PharmaPubs). <strong>IPI</strong> offers you a single<br />

source platform of communication among<br />

the professional business community of<br />

the life science industry.<br />

The current situation…<br />

On the face of it, while the global economy<br />

appears to be mired in a deepening crisis<br />

driven firstly by a lack of credit, followed by a<br />

banking crisis, collapsing residential and<br />

commercial properties markets, a sharp<br />

reversal in the demand for oil and<br />

commodities, dire predictions of an extended<br />

recession or even a depression and a general<br />

lack of investment confidence, the outlook,<br />

with interest rates at near record lows, heralds<br />

a period of deflation.<br />

In the last quarter of 2008, financial markets<br />

showed extreme volatility marked by a sharp<br />

fall in market capitalisation across most<br />

industrial sectors in anticipation of a<br />

prolonged global recession. The risk of falling<br />

interest in the face of unsustainable price<br />

increases threatened stagflation – a stagnant<br />

economy coupled with hyperinflation. Was<br />

there any justification to be just a tad more<br />

optimistic?<br />

By early December 2008, markets had<br />

already fallen to their lowest level of the year<br />

as stock prices dipped even lower than the<br />

previous low point of 31st March 2008.<br />

However, among 350 stock prices that make<br />

up the Financial Times Stock Exchange (FTSE<br />

350) index, only pharmaceuticals and non-life<br />

insurance have consistently bucked the<br />

downtrend. Could this be an indication that<br />

after several years when the pharmaceutical<br />

sector had been shunned by investors,<br />

confidence has begun to return to this<br />

defensive sector? Investor perception of risk<br />

might also translate into better returns from<br />

companies active in the non-life insurance<br />

sector where returns are more favourable.<br />

The Axeman Cometh – choppers to the<br />

rescue…<br />

I’m off to India next week and I have packed<br />

The Axeman Cometh by John Farris to read<br />

once again on the plane. Just as in the early<br />

2000s, 2008 has been a year of bear market<br />

conditions, with little signs of economic<br />

recovery. It goes without saying that the<br />

human population needs food and<br />

medicines regardless of the economic<br />

climate. However, the healthcare business<br />

may not be recession-proof. David Brennan,<br />

CEO of AstraZeneca, speaking at a<br />

Financial Times conference in London,<br />

warned that recession will be “deep and<br />

long”, indeed impacting healthcare,<br />

including the pharmaceutical industry. With<br />

drug patent expiries, and a shortage of new<br />

blockbuster drugs to fill the void, we can<br />

expect a further round of cutbacks, layoffs<br />

and other restructuring efforts aimed at costcontainment<br />

and improved efficiency within<br />

the industry. In the meantime, government<br />

initiatives aimed at stimulating the economy,<br />

with talk of “quantitative easing” on top of<br />

rapidly falling interest rates in an effort to<br />

stimulate the economy, amount to little more<br />

than printing lots of cheap money to shower<br />

the population from a fleet of helicopters<br />

circulating overhead. This could work if<br />

there was anything worthwhile to spend it on<br />

after manufacturing has been savaged.<br />

To recap - in 2008, Merck cut 8,400 jobs, but<br />

we saw major cuts at Abbott, AstraZeneca,<br />

Schering-Plough, UCB Pharma, Wyeth and<br />

at many smaller drug companies as well. In<br />

February 2009, we hear unconfirmed reports<br />

in the Daily Telegraph and Observer that<br />

GSK is preparing to lay off between 6,000<br />

and 10,000 workers. While no<br />

announcements have been made yet, the<br />

cuts are likely to hit two segments in<br />

particular – sales reps and R&D. What lies<br />

behind pharma layoffs is well-known – like<br />

so many of its rivals, GSK is facing patent<br />

expirations in the coming years, which<br />

means increased competition from generic<br />

drug-makers. The company has already cut<br />

1,800 pharma reps and 1,200 R&D workers,<br />

and shut down two plants that produce<br />

several drugs that are going off patent. The<br />

French GSK National Works council was<br />

informed of massive redundancies (848) in<br />

France – 37 people will face redundancy at<br />

the R&D centre in Les Ulis, 13 will leave the<br />

company at the Notre Dame de Bondeville<br />

plant and 798 people will leave the Evreux<br />

(Normandy) production plant by 2012. This<br />

new round of layoffs is part of CEO Andrew<br />

Witty's ongoing efforts to reorganise and<br />

diversify GSK to prepare for tough years<br />

ahead. Witty wants GSK to rely less on<br />

blockbuster drugs and more on a broad<br />

range of smaller drugs, which would protect<br />

the company from repeating the patent-loss<br />

scramble in the future.<br />

Mergers and acquisitions, consolidation …<br />

We have heard about the mega-merger of<br />

Pfizer and Wyeth. When any two megacompanies<br />

get together, something has to<br />

go – who will stand to lose most? There’s<br />

already one casualty: Crucell, the Dutch<br />

biotech company producing vaccines.<br />

Wyeth had been negotiating to buy Crucell,<br />

and since the merger, Wyeth has bailed out<br />

of those talks leaving Crucell’s stock to tank.<br />

Pfizer then announced that it planned to cut<br />

a further 10 percent of its workforce, or 7,800<br />

jobs. Analysts predict that the merger will be<br />

more likely to hit Wyeth staff numbers harder<br />

than Pfizer. To make the deal profitable, this<br />

would require deep cuts in the workforce.<br />

In February, Novartis sold US $5 billion<br />

worth from a bond sale, stocking the drugmaker’s<br />

coffers for “financial flexibility”. This<br />

was followed by Roche raising US $16 billion<br />

from a bond sale, which was the largest US<br />

dollar-denominated corporate bond sale<br />

ever. This may mean that Roche is that<br />

much closer to a takeover of Genentech.<br />

Roche’s decision to bypass banks in favour<br />

6 <strong>IPI</strong> www.ipimedia.com


of the bond markets says a couple other<br />

things, too: that the Swiss pharma wasn't<br />

confident that the troubled banking system<br />

could come through to finance the US $42<br />

billion buyout – but it is confident of<br />

eventually sealing a deal with Genentech.<br />

Otherwise, why risk paying high interest on<br />

billions in bonds? On 24th February,<br />

Bernadette Tansey reported in the San<br />

Francisco Chronicle that the unanimous,<br />

three-member committee forming<br />

Genetech’s Board formally advised non-<br />

Roche investors, who hold about 44 percent<br />

of Genentech shares, to refuse Roche's offer<br />

to take the prospering biotechnology<br />

company through the hostile takeover bid<br />

that Roche launched on 9th February - the<br />

Genentech committee had told Roche it<br />

would consider a sale at US $112 a share.<br />

Genentech argues that its chest of cancer<br />

drugs and potential new medicines offer<br />

investors the prospect of significant future<br />

growth as an independent company. But its<br />

value would be even greater as an<br />

acquisition for Roche, which already draws a<br />

substantial share of its revenues from its<br />

right to market Genentech's blockbuster<br />

cancer drugs outside the United States. By<br />

acquiring full control of Genentech, Roche<br />

would not only gain the US revenues for<br />

those drugs, but would also avoid having to<br />

renegotiate its right to sell the medicines<br />

abroad when that option expires in 2015.<br />

More recently there have been reports by the<br />

Wall Street Journal that GSK is in the early<br />

stages of discussions to acquire Piramal<br />

Healthcare for around US $1.5 billion,<br />

although these reports have been denied by<br />

the Chairman of Piramal, Mr Ajay Piramal.<br />

GSK already has a big presence in India with<br />

a sales force as large as that of Piramal;<br />

although they may not require that kind of a<br />

field force, in terms of products this would<br />

increase market share. Also, GSK may not<br />

require Piramal’s contract manufacturing<br />

facilities so there may well be some<br />

divestment. However, there are lots of<br />

question marks and as of now Mr Ajay<br />

Piramal has clearly denied the reports and<br />

has said that the news is totally unfounded<br />

–so one will need to wait and watch.<br />

All change - musical chairs…<br />

Deirdre Connelly, Eli Lilly’s President, has<br />

jumped ship to join GSK as President of its<br />

North American Pharmaceuticals. From a<br />

sales rep in 1983 to executive director of HR,<br />

then leading the entire US operation of Lilly’s<br />

US affiliate, she joined GSK reporting<br />

directly to Andrew Witty. Connelly replaced<br />

Chris Viehbacher who left to become CEO of<br />

Sanofi-Aventis. Not to be outdone, soon<br />

after Glaxo announced Connelly's<br />

appointment, Lilly tapped Enrique Conterno<br />

to take her place. Conterno had been Lilly's<br />

senior vice president of healthcare<br />

professional markets in the US.<br />

The future seems bright……<br />

According to IMS data, the global pharma<br />

market is still growing in terms of sales<br />

revenue and is forecast to hit US $820 billion<br />

in 2009, while the annual rate of growth may<br />

be levelling off at about 5 percent. Whilst the<br />

US is still the largest market, this growth is<br />

coming from the “pharmamerging” markets<br />

– the so called BRIC markets of Brazil,<br />

Russia, India, and China, plus South Korea,<br />

Turkey and Mexico. Apart from that,<br />

innovation (for example, innovation in<br />

oncology, biotech products and specialist<br />

products) is driving the product level growth<br />

in 2009 as well as an increase in generics.<br />

Novo Nordisk increased operating profit by<br />

38% in 2008 - performance driven by sales of<br />

modern insulins and gross margin<br />

improvement. The dividend is to be<br />

increased by 33%. Lars Rebien Sørensen,<br />

president and CEO, said: "We are satisfied<br />

with the solid business results achieved in<br />

2008 driven by the continued penetration of<br />

our modern insulins in all key markets.<br />

Despite the general economic downturn we<br />

still expect double-digit growth in both sales<br />

and operating profit for 2009 and we are<br />

increasing our long-term financial targets."<br />

Eli Lilly and Daiichi Sankyo have won<br />

European approval for their blood thinner<br />

Prasugrel, one of the most closely watched<br />

therapies in late-stage development.<br />

Regulators gave a green light to use the<br />

medicine on patients who had stents put in to<br />

clear arteries near the heart. The Wall Street<br />

Journal's health blog was quick to note that<br />

this is just one of many examples where<br />

Europe has led the US when it comes to new<br />

approvals. The drug will be called Efient in<br />

Europe, where it will go up against the<br />

blockbuster Plavix, which is marketed by<br />

Bristol-Myers Squibb and Sanofi-Aventis.<br />

Analysts are expecting the FDA to follow up<br />

with approval in the US following an expert<br />

panel's unanimous recommendation.<br />

Having long been regarded as an<br />

unattractive market, vaccines have reemerged<br />

as a successful growth driver for<br />

Big Pharma. The launch and rapid uptake of<br />

novel, high-price products such as Wyeth's<br />

Prevnar or Merck & Co's Gardasil, along with<br />

the emergence of novel vaccine<br />

technologies and favourable legislation, have<br />

brought vaccines back into the main focus of<br />

pharmaceutical and biotech companies.<br />

Schering-Plough will provide the World<br />

Health Organization (WHO) with access to<br />

pandemic influenza vaccine manufacturing<br />

technology in developing countries. The<br />

WHO may also give sub-licenses to firms<br />

operating in various developing countries.<br />

The agreement, which was signed by<br />

Nobilon, the company’s human vaccine<br />

business unit, grants the organisation a nonexclusive<br />

license to develop, register,<br />

manufacture, use, and sell seasonal and<br />

pandemic live, attenuated, influenza<br />

vaccines (LAIV), produced on embryonated<br />

chicken eggs. Egg-based LAIV technology<br />

is specifically considered attractive for this<br />

purpose, because the manufacturing<br />

technology process is easier to transfer,<br />

capital investment is lower, and yields are<br />

higher as compared to inactivated influenza<br />

vaccines, according to Schering-Plough.<br />

Keep them rolling…<br />

Our thanks go to all the authors of the<br />

interesting and informative articles in this<br />

issue of <strong>IPI</strong>. In future issues, we plan to<br />

review the drug discovery market, drug<br />

delivery technologies, new developments in<br />

biotech, outsourced manufacturing and<br />

clinical research developments. Enjoy this<br />

issue and we look forward to your comments<br />

and articles.<br />

Patricia Lobo, MSc, PhD<br />

As an independent management Consultant<br />

for Life Science Business Solutions (LSBS), I<br />

am honoured to be invited by Pharma<br />

Publications to be the Editor of International<br />

Pharmaceutical Industry (<strong>IPI</strong>).<br />

I have worked in the pharma sector for<br />

over 30 years after graduating in Chemistry,<br />

Microbiology and Biochemistry and gaining<br />

an external PhD in Biochemical<br />

Pharmacology and a business training<br />

course. My industrial career in the UK led<br />

from the QC department of a generics CMO<br />

to the R&D department of GD Searle (now<br />

Pfizer), followed by working as a CRA with<br />

Stiefel Laboratories, as Senior Clinical<br />

Research Scientist with Farmitalia Carlo Erba<br />

(now Pfizer), Oncology Business Unit<br />

Manager with Schering Plough, and for<br />

nearly 17 years as a Management Consultant<br />

for Technomark and RSA Consulting Ltd.<br />

There I have supported international Life<br />

Science clients with assignments in<br />

manufacturing, clinical research,<br />

development and marketing, providing<br />

specialised advice in drug development,<br />

outsourcing, clinical research or<br />

manufacturing, due diligence, strategic<br />

planning and marketing, including advice to<br />

corporate finance and equity organisations<br />

on M&As, JVs, alliances and investment. I<br />

have published over 20 scientific articles,<br />

organised a major conference and exhibition<br />

in London, set up a database of Contract<br />

Manufacturers,<br />

Email: patricia.lobo@aol.com<br />

<strong>IPI</strong> 7


<strong>IPI</strong><br />

America’s Pharmaceutical Research<br />

Companies: Improving Patient and<br />

Practitioner Knowledge about<br />

Prescription Medicines<br />

When it comes to providing patients with<br />

excellent healthcare, there is one essential<br />

ingredient that everyone in the healthcare<br />

delivery chain can subscribe to: informed,<br />

knowledgeable patients, active in their own<br />

treatment, make the best patients. Such<br />

patients are better able to understand their<br />

condition, the treatment and medical<br />

options proposed by healthcare<br />

practitioners, and are also better able to<br />

commit themselves to following through on<br />

treatments and making lifestyle and other<br />

changes that may be necessary to become<br />

healthy and stay healthy. In short, one sure<br />

way to improve the quality and utility of<br />

healthcare across the board is to empower<br />

patients with information and help them to<br />

be true partners in their own care.<br />

Fortunately, we live in an era when more<br />

information about disease, medicines and<br />

treatment options is available to both<br />

patients and medical practitioners than ever<br />

before. As a result, patients can be better<br />

advocates for their own care, and healthcare<br />

professionals can be more knowledgeable<br />

both about a patient’s conditions and also<br />

the latest, best treatment options.<br />

At the same time, however, the sheer<br />

volume of information now available – for<br />

example, via the internet – also poses a<br />

challenge to patients and healthcare<br />

professionals alike. While patients and<br />

healthcare professionals need more<br />

information about health, disease and<br />

available treatments, they also need that<br />

information to be accurate and reliable for it<br />

to be truly useful. Ideally, then, patients and<br />

healthcare practitioners need to be able to<br />

quickly and effectively sort through multiple<br />

and sometimes contradictory – though<br />

seemingly credible – sources of information<br />

in order to find answers both on diseases<br />

and their effects as well as the best<br />

treatment options. That is a daunting task for<br />

healthy healthcare consumers, and one that<br />

is all the more difficult when the consumer is<br />

also a patient confronting a serious<br />

condition or disease.<br />

As the makers of innovative medicines,<br />

America’s pharmaceutical research<br />

companies understand that patients and<br />

practitioners need reliable information so<br />

that they can make good healthcare<br />

decisions. Patients and practitioners must<br />

be able to work together to help ensure that<br />

patients get the right treatment for the right<br />

condition at the right time. Knowledge –<br />

about a disease and treatment options – is<br />

the key to ensure this critical formulation<br />

comes together for each patient.<br />

Clearly, medicines are playing an<br />

increasingly important role in healthcare.<br />

The right medicine to the right patient at the<br />

right time can not only help a patient avoid a<br />

disease or treat a disease, it can also help<br />

minimise other health issues – from costly<br />

hospitalisations and surgeries to the<br />

“Patients and practitioners<br />

must be able to work<br />

together to help ensure<br />

that patients get the right<br />

treatment for the right<br />

condition at the right time”<br />

unintended health consequences of a failure<br />

to seek treatment. The cost of avoidable and<br />

treatable chronic diseases to both patients’<br />

health and to the economy, for instance,<br />

exemplifies how important it is for patients to<br />

consult and engage practitioners and for<br />

practitioners to have broad knowledge<br />

about both disease and treatment options.<br />

Pharmaceutical advertising and<br />

marketing can and must play an important<br />

part in the provision of critical information to<br />

both patients and practitioners. America’s<br />

pharmaceutical research companies<br />

recognise both this important function of<br />

their marketing but also their obligation to<br />

help meet the information needs of patients<br />

and practitioners. This awareness has<br />

resulted in a growing commitment by<br />

America’s pharmaceutical research<br />

companies to improve the quality of their<br />

marketing efforts and also to enhance their<br />

interactions with healthcare practitioners by<br />

focusing on professionalism and<br />

transparency.<br />

In an effort to better meet the information<br />

and knowledge needs of both patients and<br />

practitioners, the Pharmaceutical Research<br />

and Manufacturers of America (PhRMA) and<br />

its member companies have embarked on a<br />

number of industry-driven initiatives<br />

designed specifically to improve the<br />

information available about the medicine<br />

they make and its appropriate uses.<br />

For example, in July 2005, PhRMA’s<br />

Board of Directors unanimously approved<br />

Guiding Principles on the Direct to<br />

Consumer Advertisements About<br />

Prescription Medicines. These guiding<br />

principles grew out of an increasing<br />

awareness of public concern over the quality<br />

of information provided in pharmaceutical<br />

direct-to-consumer advertising (DTC). The<br />

resulting principles (for more information<br />

visit: http://www.phrma.org/dtc) encourage<br />

the communication of accurate and fairly<br />

balanced information about medicines and<br />

help ensure that DTC advertising remains an<br />

important and powerful tool to help educate<br />

patients while at the same time addressing<br />

many of the concerns expressed about DTC<br />

advertising in recent years.<br />

These principles also come from an<br />

understanding – bolstered by many surveys<br />

and studies – demonstrating that patients<br />

knowledgeable about both potential<br />

diseases and possible treatments are more<br />

likely to ask their physician’s advice and be<br />

more active in protecting and improving their<br />

own health. The principles affirm that DTC<br />

advertisements are created to help inform<br />

and empower consumers and to make them<br />

aware of new medicines and their benefits<br />

as well as risks. Additionally, marketing<br />

should be designed to promote increased<br />

communication between doctors and<br />

8 <strong>IPI</strong><br />

www.ipimedia.com


Advertisement<br />

HELLO<br />

I am Karl M. Eckl,<br />

In 2000, we specialised in the type of Phase I and IIa Clinical Studies which<br />

are critical and often delay clinical development because of difficult recruitment<br />

conditions. These studies are:<br />

• PK studies in the target population (e.g. Oncology Patients)<br />

• PK studies in patient populations used as pharmacological models like<br />

patients with renal and hepatic impairment<br />

• Proof of Concept studies in the target population (phase IIa) to speed up<br />

strategic decisions in clinical development of new medicinal products<br />

Our expertise lies in:<br />

Cardiovascular<br />

CNS (including psychiatry)<br />

Endocrinology<br />

Infection disease<br />

Asthma and pulmonology<br />

Oncology<br />

Liver disease (alcohol and toxic cirrhosis<br />

and hepatitis B and C)<br />

Kidney disease (nephrology and urology)<br />

Gastroenterology<br />

Postmenopausal women<br />

Elderly population<br />

Gynaecology<br />

Rheumatoide arthritis<br />

Male and female healthy subjects<br />

Haematology<br />

Ophtalmology<br />

Pain patients<br />

Surgery and neurosurgery<br />

Why are these studies so difficult to perform?<br />

• No therapeutic benefit for patients increase reluctance to participate in such a<br />

study.Therefore it is necessary to have a panel of such patients whenever possible.<br />

• Reproducible boundary conditions are difficult to achieve in normal hospital<br />

settings. Therefore our own clinical sites are exclusively used for clinical<br />

research.<br />

I set up INNOPHAR to provide you with:<br />

• Patient populations which are the most critical ones to recruit for phase I and<br />

IIa studies (e.g. renal or hepatic impaired or oncology patients)<br />

• High quality services of our own clinical cites specifically tailored for these<br />

types of clinical studies<br />

• The service directly and not through any sub contractor<br />

• Own patient panels, we perform these studies frequently<br />

• Expertise, as phase I and IIa studies in these types of patients are our core business<br />

• We recruit patients in huge polyclinics which cover patient data from 50,000<br />

patients or more each. We co-operate with 5 – 10 policlinics, and as a result we<br />

can give exact time windows for recruitment, which is a rare thing in this business.<br />

Our Formula for success can be yours.<br />

Just contact me or my colleagues at:<br />

Dr. Karl M. Eckl<br />

Managing Director<br />

INNOPHAR GmbH<br />

INNOPHAR GmbH<br />

Innovative Pharma Research<br />

Eggersberg 4A. D-94375 Stallwang. Germany<br />

Tel: +49 (0) 9964 6010216<br />

Fax: +49 (0) 9964 6010217<br />

E-Mail: contact@ipr-ee.com<br />

www.ipr-ee.com


<strong>IPI</strong><br />

patients about existing conditions as well as<br />

other potential health issues. Further, the<br />

principles recognise that marketing and<br />

advertising can play an important role in<br />

helping to de-stigmatise a disease or<br />

condition and prompt patients to seek<br />

needed treatment.<br />

The principles, among other goals,<br />

encourage America’s pharmaceutical<br />

research companies to:<br />

• Spend appropriate time educating<br />

healthcare professionals about a new<br />

medicine before it is advertised to<br />

patients;<br />

• Include information about help for the<br />

uninsured and the under-insured in its<br />

commutations, where feasible;<br />

• Respect the seriousness of health<br />

conditions being advertised in DTC<br />

advertisements; and, critically,<br />

• Provide risk and safety information in DTC<br />

advertisements that is designed to<br />

achieve a balanced presentation of both<br />

risks and benefits associated with a<br />

medication.<br />

In addition to their DTC advertising initiative,<br />

PhRMA and its member companies have<br />

also recently focused on efforts to improve<br />

the interactions between healthcare<br />

professionals and the representatives of<br />

pharmaceutical research manufacturers. In<br />

July 2008, PhRMA introduced its revised<br />

voluntary code governing these<br />

relationships(http://www.phrma.org/news_ro<br />

om/press_releases/phrma_code_reinforces<br />

_commitment_to_responsible_interactions_<br />

with_healthcare_professionals/).<br />

The code specifically seeks to help<br />

ensure that healthcare professionals have<br />

access to the latest, most accurate<br />

information available regarding prescription<br />

medicines, while making interactions<br />

between physicians and company<br />

representatives more professional and<br />

transparent.<br />

Specifically, the code promotes<br />

communications that:<br />

• Inform healthcare professionals about the<br />

benefits and risks of biopharmaceutical<br />

medicines to help advance appropriate<br />

patient use;<br />

• Provide scientific and educational<br />

information;<br />

• Support medical research and education;<br />

and,<br />

• Provide feedback and advice about<br />

medicines and how they are being used<br />

by both manufacturers and healthcare<br />

professionals.<br />

The code also contains provisions that<br />

address provision of educational grants and<br />

continuing medical education by<br />

pharmaceutical research companies.<br />

Importantly, these voluntary initiatives by<br />

pharmaceutical research manufacturers are<br />

the result of PhRMA member companies<br />

recognising the importance to both patients<br />

and healthcare professionals of accurate<br />

and balanced information about medicines.<br />

The initiatives are also the result of<br />

pharmaceutical research companies<br />

listening to and addressing many of the<br />

concerns of both patients and practitioners<br />

about the content and quality of industry<br />

marketing practices. In the end, however,<br />

what is most important is that patients and<br />

practitioners have access to the best<br />

information about diseases, medicines and<br />

available treatment options so that they can<br />

work together to protect and improve a<br />

patient’s health and, in many instances,<br />

pharmaceutical research companies are in<br />

the best position to provide credible, needed<br />

information about the risks and benefits of<br />

the medicines they make ■<br />

Billy Tauzin was named president and chief<br />

executive officer of the Pharmaceutical<br />

Research and Manufacturers of America<br />

(PhRMA) in January 2005 and immediately<br />

took up two of the most important causes of<br />

his career. Billy Tauzin began his public<br />

service career in the Louisiana State<br />

Legislature where he served in a variety of<br />

distinguished posts such as Chairman of<br />

the House Natural Resources Committee<br />

and Chief Administration Floor Leader. He<br />

was chosen twice as one of Louisiana’s ‘Ten<br />

Best Legislators’.<br />

Email: info@phrma.org<br />

10 <strong>IPI</strong>


<strong>IPI</strong><br />

The 2008 Revised Declaration of Helsinki<br />

and Proposed Solutions for Research<br />

Ethics in Globalised Clinical Trials<br />

In October 2008 the World Medical<br />

Association (WMA) once again revised the<br />

Declaration of Helsinki (DoH), this time at<br />

its General Assembly Meeting in Seoul,<br />

South Korea. While the WMA insisted that<br />

this revision would be largely for editorial<br />

purposes, the result is a significantly altered<br />

'Ethical Principles for Medical Research<br />

Involving Human Subjects’, the current<br />

subtitle of the DoH. The revision contains<br />

both ‘editorial’ changes, including a revised<br />

use of wording in places and the integration<br />

of ‘explanatory notes’ into the main text, as<br />

well as the addition of new ‘principles’ and a<br />

greater emphasis on the primacy of the DoH<br />

Revised Ethical Principles<br />

Significant revisions appear throughout the<br />

2008 DoH. The following listing groups the<br />

principle revisions according to the thinking<br />

within the WMA that informed the revision.<br />

1. An emphasis on the individual research<br />

participant (‘subject’) whose wellbeing<br />

‘must take precedence over all other<br />

interests' (paragraph 8) and (when<br />

competent) must ‘freely agree (consent) –<br />

without exception – to the<br />

research’(paragraph 22).<br />

2. Greater emphasis on transparency in<br />

clinical research, including (a) specified<br />

information to be included in the protocol:<br />

‘information regarding funding, sponsors,<br />

institutional affiliations, other potential<br />

conflicts of interest, incentives for subjects<br />

and provisions for treating and/or<br />

compensating subjects who are harmed<br />

as a consequence of participation in the<br />

research study' (paragraph 14) and (b)<br />

specific information and methods for<br />

delivering patient information / informed<br />

consent (paragraph 24), and (c) a<br />

requirement to register publically ‘every<br />

clinical trial’.<br />

3. A restatement of the WMA position on the<br />

use of control arms in medical research,<br />

specifically indicating two exceptions<br />

where the use of placebos is allowed: (a)<br />

above other national and international<br />

ethical and legal requirements. The WMA<br />

was clearly aiming at a text to strongly<br />

impact both the international research ethics<br />

discussion and real-time practical<br />

requirements for medical research,<br />

including (if not, primarily) clinical trials.<br />

More recently, on 19 February 2009, the<br />

New England Journal of Medicine published<br />

an article entitled 'Ethical and Scientific<br />

Implications of the Globalization of Clinical<br />

Research’ in which a group of clinical<br />

triallists from the Duke University School of<br />

Medicine and the University of North<br />

‘where no current proven intervention<br />

exists’ and (b) 'where for compelling and<br />

scientifically sound methodological<br />

reasons’ in which research participants will<br />

not be exposed ‘to any risk of serious or<br />

irreversible harm’ (paragraph 32).<br />

4. A stronger position on what research<br />

participants are ‘entitled' to at the end of<br />

studies: (a) ‘to be informed of the outcome<br />

of the study' and (b) ‘to share any benefits<br />

that result from [the study]’ (paragraph<br />

33). The specific entitlement to ‘post-study<br />

access by study subjects to interventions<br />

identified as beneficial in the study or<br />

access to other appropriate care or<br />

benefits’ needs to be stated in the protocol<br />

(paragraph 14).<br />

5. A new concern with the role of ‘community’<br />

along with ‘populations’ (introduced in<br />

2000) insisting that research must be<br />

‘responsive to the health needs and<br />

priorities of this [wherein the research<br />

takes place] population or community, and<br />

if there is a reasonable likelihood that this<br />

population or community stands to benefit<br />

from the results of the research’<br />

(paragraph 17). Importantly, paragraph 22<br />

makes it clear that the individual’s interests<br />

and decisions take precedence over those<br />

of the community.<br />

Carolina School of Medicine produced a<br />

commentary raising the question: ‘To what<br />

extent should people in developing<br />

countries be enrolled in clinical trials?’ The<br />

article by US clinical researchers challenges<br />

the ethical and scientific value of clinical<br />

trials in places such as Central and South<br />

America, Eastern Europe, Southeast Asia,<br />

the Middle East, Africa, China, and India.<br />

Although they (Glickman et al.) reference the<br />

current 2008 version of the DoH, they<br />

indirectly quote the now debunked 2000-<br />

2004 version in an overall argument that<br />

says greater clarity of ethical principles is<br />

needed, greater quality control regarding<br />

clinical trial data and its geographic<br />

relevance needs to be implemented, and<br />

more government oversight is required.<br />

Since the recent and, oftentimes, heated<br />

discussion on revising the DoH began at the<br />

WMA General Assembly in Hamburg in<br />

November 1997, the WMA has repeatedly<br />

emphasised its wish that in revising the DoH<br />

12 <strong>IPI</strong><br />

www.ipimedia.com


<strong>IPI</strong><br />

the needs of research participants and<br />

researchers in ‘developing countries’ are<br />

being strengthened in relation to the<br />

perhaps real or perceived threats coming<br />

from places like the United States and the<br />

European Union, particularly with regard to<br />

commercially sponsored clinical trials. At the<br />

same time, as recently reported in this<br />

journal, leaders in the research ethics<br />

communities in these countries did not<br />

always feel that the WMA consultation<br />

process or resulting text well represented<br />

their values and concerns. Still there remain<br />

strong parallels between the concerns with<br />

the globalisation of clinical trials as<br />

expressed by the WMA, as well as by<br />

groups such as Glickman and others. In<br />

general, this is a concern that the<br />

motivations of greed, profit, and downright<br />

indifference to others does (and will<br />

continue to) allow for clinical trials that are in<br />

the interest of the powerful at the expense of<br />

the weak.<br />

Instead of refurbishing its own 'ethical<br />

principles' or adopting new ethical principles<br />

from others, as the WMA does in the DoH,<br />

Glickman and others have identified a<br />

number of issues with clinical trials in<br />

‘developing countries’ and proposed<br />

‘solutions’. Many of the issues overlap those<br />

addressed by the WMA in the DoH,<br />

including research participant selection,<br />

clinical trial transparency, the role of ethics<br />

committees/IRBs, commercial interests, and<br />

confidentiality. Glickman and others also<br />

include the issues of regulatory oversight,<br />

investigator training, a lack of<br />

pharmacogenomic information, and<br />

payments to research participants. For<br />

Glickman these issues present problems<br />

that need to be addressed, even resolved, if<br />

the current trend of increased clinical trial<br />

data from outside the US can justify the<br />

ethical and scientific confidence in the data<br />

gathered in clinical trials and used to claim<br />

new health benefits by pharmaceutical and<br />

device products. Glickman’s group<br />

proposes several concrete steps to be taken<br />

in order to provide more confidence in<br />

clinical trials carried on outside the bastions<br />

of ‘developed countries’.<br />

Solutions to the Globalisation of Clinical<br />

Trials<br />

Listed here in paraphrase are selected<br />

solutions to the ethical and scientific issues<br />

raised by clinical trials in developing<br />

countries, as proposed by Glickman and<br />

others recently in the New England Journal<br />

of Medicine:<br />

1. targeted enrolment of trial populations<br />

based on geographic region and a<br />

market match for the intended product;<br />

2. the publication of ‘all clinical trial data’<br />

ensuring the rights of investigators<br />

globally to publish independently of<br />

sponsors;<br />

3. develop a formal mechanism that allows<br />

for ‘sharing regulatory oversight’ of<br />

<strong>IPI</strong> 13


<strong>IPI</strong><br />

clinical trials globally and a public registry<br />

of ethics committees / IRBs globally;<br />

4. create training programmes for<br />

investigators in clinical research and<br />

ethics;<br />

5. use the US FDA Voluntary Genomic Data<br />

Submissions programme globally across<br />

other regulatory agencies;<br />

6. increase the use of centralised ethical<br />

review procedures;<br />

7. adopt standard contract language and<br />

confidentiality agreements for clinical<br />

trials.<br />

Glickman concludes the article indicating<br />

that a single listing of fixes to the challenges<br />

of ethics and science in today’s international<br />

clinical trials market place is unlikely to bring<br />

satisfaction in and of itself: ‘Long-term<br />

solutions to problems arising from the<br />

globalization of clinical research will require<br />

input from stakeholders in academia,<br />

industry, and regulatory agencies around<br />

the world.’ The authors, however, appear to<br />

still want to limit the discussion to those with<br />

the money, power, and/or authority: they fail<br />

to recognise the contribution that<br />

representatives from ethics<br />

committees/IRBs and, perhaps most<br />

importantly, representatives of patient and<br />

consumer organisations can make to the<br />

efficiency, as well as the ethical and<br />

scientific understanding, of clinical research.<br />

Without input from a broader stakeholder<br />

group, the engagement in globalised clinical<br />

trials is highly unlikely to get either the<br />

principles or the solutions, let alone the<br />

practices, right. At this time when more is<br />

being called for, even more is needed.<br />

Indeed the structure of the international<br />

discussion, alongside how and to what<br />

extent contributions may be made to that<br />

discussion, is a major issue that haunts the<br />

WMA in defending its revised DoH. The<br />

process for the present revision of the DoH<br />

began in May 2007 with the establishment of<br />

a Working Group composed of<br />

representatives from the national medical<br />

associations of Brazil, Germany, Japan,<br />

South Africa, and Sweden. From the outset,<br />

the primary purpose of this revision process<br />

was termed 'editorial': the WMA was largely<br />

interested in integrating the Notes of<br />

Clarification to paragraphs 29 and 30 of the<br />

2000 version that were added in 2002 and<br />

2004. Major issues were not immediately in<br />

the remit of the Working Party, and it was<br />

explicitly barred from addressing the<br />

fundamental structure and arrangement of<br />

the text. The WMA felt that the Notes of<br />

Clarification were not clearly understood in<br />

relation to the texts they were intended to<br />

clarify.<br />

The WMA also proposed to use this<br />

opportunity to clarify other issues in Helsinki<br />

and to address specific issues that recently<br />

appeared to be of importance and in need<br />

of global clarification. Just why certain<br />

issues were chosen for inclusion (for<br />

example, the registration of clinical trials)<br />

and others were set aside (for example,<br />

research on healthy volunteers) has not<br />

been publicly disclosed by the WMA,<br />

though it may be that there was a certain felt<br />

political advantage within the organisation to<br />

adapt recommendations from other<br />

organisations. Between May 2007 and<br />

September 2008, the WMA invited two<br />

rounds of comments on the DoH and<br />

hosted three workshops to discuss the<br />

issues and proposed changes. No public<br />

reports of the comments received nor of the<br />

workshops were prepared and, although<br />

there have been calls for transparency with<br />

regard to the WMA process for revising<br />

Helsinki, the WMA has decided not to make<br />

public the comments received or reports of<br />

the workshops. While the pharmaceutical<br />

industry seems to have been well integrated<br />

into the process through the participation of<br />

the International Federation of<br />

Pharmaceutical Manufacturers and<br />

Associations (IFPMA) as well as the<br />

participation of individual companies, there<br />

does not appear to have been a structured<br />

discussion with representatives from such<br />

groups as ethics committees or patient<br />

organisations.<br />

Glickman and others have also run up<br />

against similar obstacles in proposing<br />

solutions to problems in globalised clinical<br />

research. Although they call for a broader<br />

discussion on the problems they see as being<br />

brought about by the globalisation of clinical<br />

trials, their proposed solutions are limited to a<br />

group of researchers all living and working<br />

within a 100 kilometre radius inside the posh<br />

American south. As they look eastward past<br />

Washington, DC and Brussels, there is a<br />

tendency to characterise the larger part of the<br />

world (of clinical trials) outside the US and the<br />

EU as ‘developing countries’. It appears all too<br />

easy not to look westward in one's own<br />

backyard of Appalachia and the vulnerable<br />

characteristics of many of the research<br />

participants that populate the clinical trials in<br />

Durham and Chapel Hill, North Carolina. It is<br />

perhaps too simple to believe that their clinical<br />

trial cohorts are socially better off than cohorts<br />

in Moscow, Mumbai, Singapore, Shanghai, or<br />

Buenos Aires. If the participants in this<br />

discussion are not attentive and listening,<br />

there is the clear and present danger of<br />

mistaking national and cultural identity for<br />

some superior position on the ladder of ethical<br />

and scientific justification. At the very least<br />

proposals for solutions to the challenges of<br />

international clinical trials require international<br />

authorship, and more than a reference listing<br />

of 62 references with only a single one drawn<br />

from a so-called developing country, China.<br />

It is now an accepted fact that clinical trials<br />

are no longer primarily confined to the ICH<br />

region that provided the background to the<br />

1996 E-6 Good Clinical Practice Guideline.<br />

The United States, Japan, and the European<br />

Union (even having more than doubled in<br />

size) no longer have any special claim to<br />

hosting clinical trials. This flattening of the<br />

clinical trials marketplace does lead to a<br />

rethinking of the key issues in research ethics,<br />

as well as the methodological and quality<br />

characteristics of clinical trial data on which we<br />

base scientific and public health decisions.<br />

Glickman and others are right to assert: ‘The<br />

future of the pharmaceutical and device<br />

industries is predicated on addressing these<br />

issues.’ This future will depend, as the WMA<br />

has seen, on a reappraisal of the ethical<br />

principles that underlie clinical research as<br />

well as on an attempt to collectively seek<br />

solutions, as Glickman and others indicate, to<br />

contemporary ethical and scientific issues in<br />

clinical trials. More importantly, however, the<br />

future of these industries, both academic and<br />

commercial, will require us to revise our<br />

practices to meet the new realities which<br />

present themselves in science, culture, and<br />

ethics ■<br />

Francis P. Crawley<br />

is the Executive<br />

Director of the Good<br />

Clinical Practice<br />

Alliance – Europe<br />

(GCPA) in Brussels,<br />

Belgium and a World<br />

Health Organization<br />

(WHO) Expert in ethics. He is the co-founder<br />

of the Strategic Initiative for Developing<br />

Capacity in Ethical Review (SIDCER). He is<br />

a past member of the UNAIDS Ethical<br />

Review Committee and currently Chairs the<br />

Ethical Review Committee at the<br />

International Network for Cancer Treatment<br />

and Research (INCTR). He has played a<br />

pivotal role in developing European and<br />

international guidance in ethics and<br />

regulation for health research.<br />

Email: fpc@gcpalliance.org<br />

14 <strong>IPI</strong>


<strong>IPI</strong><br />

e-Networking in the<br />

Drug Discovery Industry<br />

The complexities of modern drug<br />

discovery often require cooperation<br />

between many different participants,<br />

sometimes located in different continents.<br />

This is particularly true in an outsourcing<br />

environment, where clinical trials,<br />

manufacturing and even preclinical<br />

research become globalised. Demands for<br />

effective communication between<br />

companies, laboratories and individuals<br />

have therefore increased enormously.<br />

Fortuitously, or possibly causatively, these<br />

demands coincide with the supply of<br />

internet-based networking tools that make<br />

geographical boundaries and time zones<br />

irrelevant. e-Networking is expanding<br />

rapidly and is here to stay.<br />

Background<br />

The pharmaceutical industry is beginning to<br />

reinvent itself as commercial pressures<br />

make the old FIPCo (fully integrated<br />

pharmaceutical company) model<br />

increasingly unsupportable. For example,<br />

patent expiries on best selling medicines<br />

over the period 2007 to 2012 will cost the top<br />

50 companies $115 billion in lost revenues<br />

(the patent cliff) (1). It is no longer possible<br />

for companies to replace these lost sales<br />

revenues by remaining isolated, so<br />

partnership models are being developed to<br />

create the fully integrated pharmaceutical<br />

network (FIPNet). This involves sharing<br />

financial risk with partners, as well as<br />

outsourcing many research and<br />

development functions that were once kept<br />

firmly in-house. Large western<br />

pharmaceutical companies are increasingly<br />

turning to high-growth economies such as<br />

China and India for support, from discovery<br />

research (e.g. medicinal chemistry and<br />

biotechnology) all the way through to<br />

manufacturing.<br />

This globalisation process has been<br />

greatly facilitated by the internet, through<br />

improved person-to-person communication<br />

(email), as well as through information<br />

portals which provide access to regulatory<br />

guidelines, research literature etc. Next<br />

generation web-based tools which use Web<br />

2.0 technology make information flow a truly<br />

two-way process by using blogs, wikis and<br />

social/business networking sites.<br />

To be successful in drug development,<br />

such tools must increase the interchange of<br />

ideas between scientists, clinicians and<br />

business people. To be effective, they must<br />

help to increase the rate of discovery,<br />

development and marketing of new<br />

medicines.<br />

We have recently reviewed developments<br />

in scientific networking from the perspective<br />

of the drug discovery scientist (2, 3). In the<br />

current article, we extend these observations<br />

to cover the broader drug development and<br />

“Next generation webbased<br />

tools which use<br />

Web 2.0 technology<br />

make information flow<br />

a truly two-way process<br />

by using blogs, wikis<br />

and social/business<br />

networking sites”<br />

marketing arena. We conclude that social<br />

and business networking technologies<br />

increasingly provide a serious, credible and<br />

essential resource for the pharmaceutical<br />

and biotechnology industries.<br />

e-Networking and Web 2.0<br />

Improvements in networking and<br />

communication technologies have given rise<br />

to new concepts, particularly that of Web<br />

2.0. Web 1.0 is the well-established internet<br />

of static web pages and downloadable<br />

content (“pull” technology). Web 2.0,<br />

however, allows direct user control (“push”<br />

technology) of content on remote web<br />

pages. The Web 2.0 concept involves<br />

“harnessing the collective intelligence” of a<br />

community (4). This does not imply a major<br />

change of technology, but is more about<br />

“teaching the old web new tricks” (5). These<br />

“new tricks” include blogs, wikis,<br />

folksonomies, RSS feeds and mashups.<br />

Blogs (web logs) contain regularly<br />

updated personal commentary, descriptions<br />

of events, diary entries etc that are hosted by<br />

individuals or organisations on their<br />

websites. Blogs are very powerful<br />

networking tools, since others may<br />

contribute interactively in the form of online<br />

conversations. Sometimes the content is<br />

delivered as audio in the form of podcasts,<br />

or video (vlogging).<br />

Wikis (Hawaiian for “fast”) are information<br />

resources that can be edited by the online<br />

community. The most famous example is<br />

Wikipedia (http://en.wikipedia.org/), an<br />

online encyclopaedia that is continually<br />

evolving as content is added, removed or<br />

modified.<br />

Folksonomies (folk taxonomies) consist<br />

of search terms provided by the online<br />

community to search the web according to<br />

individual preferences. This feature<br />

distinguishes folksomonies from the more<br />

controlled vocabularies used by search<br />

engines to find information on the internet.<br />

RSS feeds (Real Simple Syndication) are<br />

a form of aggregator that automatically pulls<br />

together updated information, such as news<br />

headlines or recent publications, and<br />

delivers the content directly to users as a<br />

single display on the web page.<br />

Mashups are applications that combine<br />

data from more than one source into a<br />

feature such as a generalised world map<br />

overlaid with specific information. An<br />

example of this is the community map for the<br />

Drug Design Resource, described in detail<br />

elsewhere (2, 3). These technologies are<br />

evolving rapidly and can be found within an<br />

increasing number of internet networking<br />

sites.<br />

Social networking<br />

Most internet users are aware of social<br />

networking sites such as Facebook,<br />

MySpace and Bebo. Because they are used<br />

16 <strong>IPI</strong><br />

www.ipimedia.com


<strong>IPI</strong><br />

mostly for social interactions among young<br />

people, there are obvious limitations to their<br />

utility in professional interactions among<br />

drug industry personnel. However, there is<br />

some activity in the drug development area<br />

as we have described in a previous article<br />

(2). Whatever the topic under discussion,<br />

they give their members access to specific<br />

networks regardless of professional status,<br />

and allow members to make new contacts<br />

almost instantaneously.<br />

Traditional means of networking, such as<br />

telephoning, sending emails or attending<br />

conferences are often much less efficient.<br />

Interestingly, the social network model has<br />

been adopted by a specialist medical<br />

imaging organisation (High Frequency<br />

Clinical & Preclinical Imaging Community) to<br />

support their Annual Convention<br />

(http://www.aium2009.org). Individual<br />

groups have been set up along the<br />

Facebook model for different specialisations<br />

(e.g. sonography or thyroid imaging). We<br />

expect to see this more widely adopted by<br />

the wider community.<br />

Lastly comes Twitter (www.twitter.com).<br />

This site hosts one-sentence<br />

thoughts/comments/links on any subject<br />

that can be followed by those with the<br />

inclination to do so. This internet version of<br />

text messaging has been used by<br />

organisations as well as individuals, and<br />

there is a surprising amount of material<br />

related to drug discovery. It remains to be<br />

seen whether this will be a passing fad, or<br />

something genuinely useful for near realtime<br />

alerts for news on science and<br />

business.<br />

Business networking<br />

Most business networking is conducted<br />

face-to-face at meetings and conferences,<br />

although cost issues have encouraged the<br />

uptake of video conferencing. The internet<br />

itself has also been used as a<br />

videoconferencing tool. For example,<br />

companies like WebEx<br />

(http://www.webex.com/) provide video and<br />

text links in a variety of configurations that<br />

allow sophisticated networking to anyone<br />

with a broadband connection. More value is<br />

being extracted from conferences by their<br />

organisers through podcasts and web<br />

seminars (webinars) that provide material for<br />

those unable to attend.<br />

Business networking sites are flourishing,<br />

particularly because of the dynamic job<br />

market and the increasing competition for<br />

business. Sites like LinkedIn, Ryze and Xing<br />

provide a more structured environment for<br />

the establishment of personal networks. We<br />

have discovered from our own experience<br />

with the Group facility within LinkedIn that<br />

these sites provide powerful networking<br />

platforms for collaboration (see<br />

http://www.linkedin.com/groups?home=&gi<br />

d=151138&trk=anet_ug_hm and<br />

http://www.linkedin.com/groups?gid=13309<br />

1&trk=hb_side_g)<br />

Drug discovery and development networks<br />

A broad group of disciplines, covering<br />

science, medicine, engineering and the law,<br />

are required to bring drugs from the<br />

laboratory to the marketplace. Expert<br />

communities centred on these disciplines<br />

can share valuable information and<br />

experience on topics of mutual interest. The<br />

following examples give a flavour of the<br />

potential applications in drug development:<br />

1) Pooling expertise on drug safety<br />

The Drug Safety Executive Council (DSEC)<br />

(http://www.drugsafetycouncil.org/index.cfm<br />

) in the USA is a network of over 1500 drug<br />

safety personnel in pharmaceutical<br />

companies. Each company is in a<br />

competitive marketplace, but decided to<br />

pool information on technologies to test<br />

drug safety because of the trust established<br />

through a networking infrastructure driven by<br />

the web. This benefits all companies without<br />

compromising proprietary information.<br />

2) Exchange of clinical information<br />

Pfizer announced a strategic partnership<br />

with Sermo (www.sermo.com) in 2007 as an<br />

early example of social networking by a large<br />

pharmaceutical company to further its<br />

business objectives. This direct online<br />

discussion with physicians is a significant<br />

departure from the normal exchange of<br />

medical information about the company’s<br />

products, be they formal conferences or<br />

sales visits to surgeries.<br />

3) Drug discovery for the developing world<br />

There is a well-known mismatch between the<br />

need for novel drugs to treat diseases<br />

endemic in the developing world and the<br />

willingness of the pharmaceutical industry to<br />

divert resources away from “western”<br />

diseases. This situation is beginning to<br />

change through partnerships between major<br />

drug companies, academia and small<br />

companies in the developing world. The<br />

internet provides an ideal tool for developing<br />

networks between these disparate<br />

organisations. One example is a<br />

coordination strategy for lead discovery for<br />

malaria, tuberculosis (TB), African sleeping<br />

sickness, Leishmaniasis and Chagas<br />

disease (6, 7). Here, the use of resources is<br />

highly coordinated within innovation<br />

networks or virtual drug discovery networks.<br />

A similar concept is provided by an Indian<br />

initiative “Open Source Drug Discovery”<br />

<strong>IPI</strong> 17


<strong>IPI</strong><br />

(http://www.osdd.net/what_is_osdd.htm),<br />

and also for TB.<br />

4) In silico drug design<br />

Alongside several other resources,<br />

Biovillage has established the specialist in<br />

silico drug design networking site - The Drug<br />

Design Resource (DDR) – employing<br />

traditional information resources enhanced<br />

by Web 2.0 technologies. The latter includes<br />

RSS feeds of the latest drug design literature<br />

as well as map-based mashups of the<br />

community. This resource is currently being<br />

expanded to provide both additional content<br />

and added connectivity (see below).<br />

The evolution of membership-driven<br />

content at The Drug Design Resource<br />

As a first step in producing an integrated<br />

suite of resources in the drug design area,<br />

we have linked a series of Web 2.0 platforms<br />

together, as shown in Figure 1. Staple<br />

information sources derived from peerreviewed<br />

journal articles, published books<br />

and conference reports are assembled<br />

through the curated DDR website<br />

(http://www.drugdesignresource.com/). The<br />

Resource is also linked directly to<br />

spontaneous blogsites, including the FBDD<br />

blogspot at www.practicalfragments.<br />

blogspot.com and the SBDD blogspot at<br />

http://blogs.informa.com/sbdd), both of<br />

which provide conduits for immediate<br />

feedback from the community. At the same<br />

time, the DDR is dynamically linked to<br />

individual entries in the professional<br />

networking site LinkedIn, providing an up-todate,<br />

curated ensemble of individual contacts<br />

and additional, person-centric information.<br />

Such an architecture provides immediate<br />

Fig. 1<br />

Expanding connectivity within the<br />

drug design community through<br />

various e-networking modalities<br />

Curation<br />

Journals<br />

Curation<br />

Blog<br />

Leaders in<br />

e-Drug Design<br />

DDR<br />

LinkedIn<br />

access to a wide variety of valuable<br />

information, ranging from the individual to<br />

the corporate. It can also bridge research<br />

taking place on different continents and<br />

within different disciplines, providing a<br />

unique platform for the exploration of<br />

relevant discovery information. It is also<br />

cumulative, with scientific content evolving in<br />

parallel with individual contributions.<br />

More importantly, it provides a ‘voice’ for<br />

its membership, in the first instance the<br />

development of a project networking facility<br />

(for more information see the Drug Design<br />

Directory at www.drugdesignresource.com).<br />

Conclusions<br />

Scientific communication and networking<br />

underpin the globalisation of the<br />

pharmaceutical industry, a trend which is<br />

being facilitated through the introduction of<br />

new web-based tools (Web 2.0). In this<br />

article, we have discussed some of the<br />

approaches to scientific networking that are<br />

relevant to drug development. The common<br />

theme is interactive participation, whether by<br />

signing up to a social or professional online<br />

network, contributing to a blog or discussion<br />

board, or creating/editing a Wiki.<br />

It has always been recognised that<br />

personal interaction is vital for seeding and<br />

developing scientific ideas. Web 2.0<br />

technologies simply expand the number of<br />

people that can participate in these<br />

conversations. What were once monologs<br />

from static Web 1.0 technology have now<br />

become ‘polylogs’. These are changing<br />

rapidly as their utility in essential<br />

professional activities – such as job hunting,<br />

opinion seeking, data sharing and<br />

collaboration – becomes evident ■<br />

Facebook<br />

Peer Review<br />

Books<br />

Peer Review<br />

Dr David Bailey is a<br />

Cambridge-based<br />

entrepreneur. Cofounder<br />

of IOTA<br />

Pharmaceuticals Ltd<br />

and BioVillage Ltd, he<br />

was previously the<br />

founding CEO of two<br />

Cambridge-based start-ups, De Novo<br />

Pharmaceuticals Ltd and Purely Proteins Ltd.<br />

Dr Bailey headed up the Molecular Sciences<br />

Department at Pfizer in Sandwich for 8 years,<br />

before becoming Vice President at the<br />

Californian biotech company Incyte<br />

Genomics. He is a Board Director of the<br />

Babraham Institute in Cambridge and<br />

consultant entrepreneur for the University of<br />

Greenwich in London.<br />

Email: david.bailey@biovillage.co.uk<br />

Dr Ed Zanders has an<br />

extensive research<br />

background in<br />

biochemistry and<br />

immunology through<br />

over thirty years’<br />

experience in<br />

academia and industry.<br />

He spent 16 years at<br />

Glaxo in the UK and Switzerland where he was<br />

a Senior Research Manager with responsibility<br />

for drug discovery programmes in<br />

inflammatory diseases. He later joined De<br />

Novo Pharmaceuticals Ltd and Purely Proteins<br />

Ltd in VP positions. In addition to co-founding<br />

BioVillage Ltd, he has also established a<br />

pharmaceutical training company,<br />

ScienceInform Ltd.<br />

Email: ed.zanders@biovillage.co.uk<br />

References<br />

1. <strong>Editorial</strong> Nature Rev Drug Disc 8: 3-7 2009<br />

2. Bailey DS and Zanders ED, Drug Discovery<br />

in the Era of Facebook – New Tools for<br />

Scientific Networking, Drug Discovery<br />

Today, 13:863-8 2008<br />

3. Bailey D and Zanders E, e-Networking and<br />

Drug Discovery. Innovations in<br />

Pharmaceutical Technology, October 2008<br />

4 .<br />

http://www.oreillynet.com/pub/a/oreilly/tim/ne<br />

ws/2005/09/30/what-is-web-20.html<br />

5. Sagotsky JA et al. Life Sciences and the<br />

web: a new era for collaboration. Molecular<br />

Systems Biology 4:201 1-10 2008<br />

6. Solomon N and Hudson A, Innovative lead<br />

discovery strategies for tropical diseases. Nature<br />

Reviews Drug Discovery 5: 941-955 2006<br />

7. Hopkins AL, Witty MJ and Nwaka S, Mission<br />

Possible. Nature 449: 166-169 2007<br />

18 <strong>IPI</strong>


<strong>IPI</strong><br />

Strategies for the formulation of poorly<br />

soluble compounds and polymorph<br />

drugs based on polyelectrolyte carriers<br />

The main challenge in the administration of<br />

poorly soluble drugs is to design oral<br />

formulations displaying a relevant<br />

bioavailability and to develop injectable<br />

systems for the parenteral application with<br />

minimised toxicity and high drug loading.<br />

The LBL-Technology® offers a strategy for<br />

enhancing the kinetic of dissolution of BCS<br />

class II drugs based on the presentation of<br />

the drug in the form of nanoparticles<br />

stabilised with layers of polyelectrolyte<br />

complexes (Fig. 1).<br />

In this way, nanosuspensions of drug<br />

substances with impaired water solubility<br />

can be produced displaying a particle size<br />

distribution in the nanometre range,<br />

modified surface charge and high drug<br />

concentrations (Fig. 2). These drug delivery<br />

systems are especially attractive for their low<br />

aggregation in further galenic steps in the<br />

development of solid dosage forms and for<br />

their fast dissolution profiles.<br />

There is a broad range of polycationic<br />

and polyanionic pharmaceutical excipients<br />

which build complexes by means of their<br />

electrostatic interaction. Polyelectrolyte<br />

complexes are amorphous carriers, which,<br />

depending on the production method and<br />

composition, present different rheological<br />

and plastic behaviours, mechanical<br />

properties and degrees of porosity (Fig. 3).<br />

These polyelectrolyte matrices are optimal<br />

scaffolds for the precipitation of polymorph<br />

drugs in the most preferred amorphous<br />

state (Fig. 4). These compositions, besides<br />

stabilising the drug in the amorphous state,<br />

also have the additional role of being<br />

appropriate filler excipients for solid dosage<br />

form development. Polyelectrolyte<br />

complexes loaded with drugs can be<br />

produced in the form of sponge-like<br />

supramolecular structures presenting an<br />

amorphous nature and with improved<br />

rheological properties, stability, solubility and<br />

later bioavailability.<br />

Fig. 1<br />

CLSM (confocal laser scanning microscopy) of a nanosuspension of a BCS class II<br />

drug stabilised with polyelectrolyte layer (left); detailed representation of the particle<br />

surface (right)<br />

Fig. 3<br />

Polarised microscopy<br />

of a sponge-like<br />

polyelectrolyte<br />

complex.<br />

Maria Gonzalez Ferreiro, MSc Pharmacy,<br />

PhD Pharmaceutical Technology (Freie<br />

Universität Berlin, Germany) is Head of<br />

Pharmaceutical Development at Capsulution<br />

Pharma AG in Berlin.<br />

Email: m.ferreiro@capsulution.com<br />

Fig. 2<br />

Particle size distribution<br />

of a nanosuspension<br />

formulated via LBL-<br />

Technology®<br />

Fig. 4<br />

X-ray powder diffraction<br />

(XRPD) of a control<br />

polymorph drug in the<br />

crystalline state (grey), a<br />

drug-free sponge-like<br />

polyelectrolyte carrier<br />

control (black) and<br />

different formulations<br />

thereof displaying the<br />

drug stabilised in the<br />

most preferred<br />

amorphous state (red,<br />

blue, green).<br />

20 <strong>IPI</strong><br />

www.ipimedia.com


Does your CRO pay<br />

enough attention<br />

to detail?<br />

Experience plays a crucial role in the field of Clinical Research.<br />

That is why after 10 years we have become the best in the business and<br />

have earned your valuable trust.<br />

Through 10 years of dedicated research in more than 70<br />

Phase II – IV Clinical Studies and 3 FDA and 1 EMEA inspections, we have<br />

developed an impeccable track record across fifteen countries in Greece,<br />

Turkey and the MENA (Middle East & North Africa) region.<br />

Our attention to detail has helped us to excel in:<br />

• Feasibility Studies<br />

• Investigator selection<br />

• Regulatory submissions<br />

• Trial management and monitoring<br />

• Project Management<br />

Because<br />

Expertise<br />

comes with<br />

Experience<br />

MEK Consulting, Berytech Technological Pole, Mar Roukos, 1107 2240<br />

Mkalles Lebanon, Tel: +961 (4) 531 372 Fax: +961 (4) 539 087<br />

Email: enquiry@mekconsulting.com www.mekconsulting.com<br />

Consulting<br />

Contract Research Organisation


<strong>IPI</strong><br />

Clinical research education<br />

& training: opportunties<br />

and issues<br />

Background<br />

Clinical trials were initiated around 1537, but<br />

came to the limelight when James Lind in<br />

1747 first introduced control groups in a<br />

study of scurvy. He later went on to become<br />

the father of clinical trials. Clinical trials<br />

started flourishing from 1800’s, and the<br />

focus of studies used to be on study design.<br />

From 1863 onwards scientists started using<br />

placebos. Randomised studies were<br />

initiated in 1923, and from 1945 the focus<br />

moved to ethical aspects of clinical trials.<br />

However, a lot has changed in the clinical<br />

research scenario since then. Today, clinical<br />

trials are conducted through a regulated<br />

approach following certain guidelines laid<br />

down by the International Conference on<br />

Harmonization (ICH), which is spearheaded<br />

by the USA, Europe and Japan.<br />

Clinical research constitutes a large<br />

sector of the healthcare industry and<br />

employs a significant percentage of the<br />

healthcare workforce. The biomedical<br />

research industry spends an estimated<br />

US$38 billion worldwide annually. Out of this,<br />

the global CRO market size is US$15 billion,<br />

growing at 16% per annum.<br />

The growing demand for clinical trial<br />

professionals has led to an increasing<br />

number of institutions offering academic<br />

programmes in clinical research. According<br />

to a 2001 CenterWatch survey, the US has<br />

slightly more than 200,000 clinical research<br />

professionals. There are 60,000 trials<br />

following the Food and Drug<br />

Administration’s guidance for GCPs and<br />

more than 40,000 GCP-trained investigators,<br />

but the number of investigators is<br />

decreasing even as the number of trials<br />

increases. According to some reports there<br />

are over 1000 global competitors in the CRO<br />

segment offering various services, and it is<br />

estimated that over 40,000 subjects<br />

participate in global clinical trials every year.<br />

Recruitment is a major stumbling block in<br />

the drug development process, and<br />

increasing staff costs mean that more and<br />

more studies will be outsourced to the<br />

emerging markets. Sponsors are looking at<br />

emerging markets to leverage the high cost<br />

of trials in the US and Europe, and to reduce<br />

time to market.<br />

Need for Education & Training<br />

The demand for clinical research<br />

professionals is rising globally and clinical<br />

trials are being outsourced from several<br />

countries, including rapidly growing<br />

emerging markets like India and China. As a<br />

result, human resource managers have a<br />

challenge in front of them to find out or<br />

develop the right quality of manpower. In<br />

addition to hiring experienced professionals,<br />

they need to consider clinical research<br />

professionals who have undertaken formal<br />

training or education in clinical research.<br />

In the developed markets target<br />

audiences for clinical research education<br />

and training are often those persons who are<br />

currently active in clinical research. However,<br />

in the emerging markets the target<br />

audiences of such training programmes are<br />

graduates and postgraduates in medicine,<br />

science, pharmacy and other related<br />

subjects without experience in clinical<br />

research. This new strategy of training fresh<br />

graduates has been adopted to build<br />

capacity, and develop a new well-trained<br />

clinical research workforce in the emerging<br />

markets.<br />

Clinical research managers /<br />

coordinators are an important part of the<br />

clinical trial process. In order to be a<br />

successful clinical trial professional the<br />

person would need to have formal training in<br />

clinical research or previous work<br />

experience in clinical trials, and a working<br />

knowledge of the clinical research industry.<br />

Programme Content<br />

A good clinical research training programme<br />

must provide knowledge of GCP (good<br />

clinical practices), and the Code of Federal<br />

Regulations or regulations of other<br />

countries, viz. EMEA, MHRA, Schedule Y of<br />

India, Anvisa of Brazil, TGA Australia, Health<br />

Canada and several other countries like<br />

China and Russia. It is important that the<br />

training of clinical research managers and<br />

coordinators emphasises the history of<br />

research regulation, guidelines and forms,<br />

codes of federal regulations, phases of<br />

clinical research, IEC / IRBs and submission<br />

requirements, informed consent process,<br />

protocol review and evaluation, project<br />

management, budget preparation and<br />

negotiation, regulatory submissions,<br />

adverse events, study drug accountability,<br />

and setting up one’s research site.<br />

It takes a lot of responsibility and<br />

experience to be a clinical research<br />

professional. It is important for the clinical<br />

research professionals to understand ethical<br />

issues involved in human research, follow<br />

regulations stringently, work effectively with<br />

other stakeholders like IEC/IRB, create and<br />

conduct informed consent, manage study<br />

data, manage adverse events, understand<br />

the community/research interface, collect<br />

high quality data, identify funding sources<br />

and prepare grant proposals, and fulfil<br />

reporting obligations.<br />

Clinical research professionals must<br />

learn the drug development process;<br />

understand global clinical research<br />

regulations, the informed consent process<br />

and the relevant regulations; help coordinate<br />

sponsor site visits; manage clinical trial<br />

protocol; and identify ethical issues in<br />

clinical research and their impact on the<br />

development of good clinical practices.<br />

Participating in audits is important for a<br />

clinical research coordinator because<br />

without the training, a clinical coordinator<br />

would not be able to perform or face the<br />

audit to high quality standards for an<br />

inspection. It is important to maintain a safe<br />

and quality environment in clinical settings to<br />

ensure the individuals participating in the<br />

clinical trials, and the future consumers, will<br />

be safe.<br />

Until about the early part of 2000, a<br />

science or pharmacy graduate could easily<br />

get a job as a clinical research coordinator<br />

www.ipimedia.com<br />

<strong>IPI</strong> 23


Does your CRO pay<br />

enough attention<br />

to detail?<br />

Experience plays a crucial role in the field of Clinical Research.<br />

That is why after 10 years we have become the best in the business and<br />

have earned your valuable trust.<br />

Through 10 years of dedicated research in more than 70<br />

Phase II – IV Clinical Studies and 3 FDA and 1 EMEA inspections, we have<br />

developed an impeccable track record across fifteen countries in Greece,<br />

Turkey and the MENA (Middle East & North Africa) region.<br />

Our attention to detail has helped us to excel in:<br />

• Feasibility Studies<br />

• Investigator selection<br />

• Regulatory submissions<br />

• Trial management and monitoring<br />

• Project Management<br />

Because<br />

Expertise<br />

comes with<br />

Experience<br />

MEK Consulting, Berytech Technological Pole, Mar Roukos, 1107 2240<br />

Mkalles Lebanon, Tel: +961 (4) 531 372 Fax: +961 (4) 539 087<br />

Email: enquiry@mekconsulting.com www.mekconsulting.com<br />

Consulting<br />

Contract Research Organisation


<strong>IPI</strong><br />

programmes and graduate & postgraduate<br />

degrees. Most of these programmes vary<br />

from country to country, and there is no<br />

standardisation of the course curriculum.<br />

There is a need for short-term specialised<br />

programmes. Although university<br />

programmes offer long-term programmes<br />

on clinical research, there is a great need for<br />

short duration specialised programmes on<br />

subjects like pharmacovigilance, drug<br />

safety, biostatistics, quality assurance in<br />

clinical research, bioequivalence &<br />

bioavailability, subject recruitment &<br />

retention, regulatory issues and several<br />

other such topics. These programmes<br />

provide more practical training to the<br />

students in the workshop environment.<br />

Challenges in Clinical Research<br />

Education & Training in the absence of a<br />

standardised curriculum there are gaps in<br />

the training programmes.<br />

There are several factors which limit the<br />

implementation of quality clinical research<br />

education and training programmes. One of<br />

the critical issues is the quality of training<br />

facilities. There is a lack of commitment in<br />

the industry and investigators to train<br />

budding clinical research professionals.<br />

Some other issues are the allocation of<br />

training budgets by companies. Most<br />

companies consider training and education<br />

to staff as an incentive rather than an<br />

investment to improve the quality of clinical<br />

trials conducted by them.<br />

Another issue which impacts training is the<br />

high cost of the large number of programmes.<br />

Most of the programmes offered have<br />

prohibitive costs, hence most students and<br />

even working professionals cannot afford<br />

these programmes.<br />

There are several other challenges which<br />

need to be addressed to implement the quality<br />

programmes, viz. who should be trained, who<br />

should provide training, course design,<br />

programme delivery mechanisms, cost, role of<br />

the industry, accreditation, what role<br />

professional associations could play in setting<br />

the benchmarks, whether there is a need for<br />

formal university programmes or whether<br />

certification / diploma programmes are able to<br />

address the needs of the industry. Several<br />

such questions are unanswered today.<br />

Like other professional education<br />

programmes, e.g. management education,<br />

there is a great need for the educational and<br />

training institutions to arrive at a common<br />

forum together and device a strategy to<br />

address the above challenges ■<br />

Kamal Shahani<br />

received his Masters<br />

in Business<br />

Administration from<br />

FORE School of<br />

Management in New<br />

Delhi and also holds<br />

Degree in Commerce<br />

and Post Graduation in International<br />

Business. He has over 20 years experience<br />

in the Indian healthcare and clinical research<br />

industry on the senior management positions<br />

and has set up and managed large<br />

healthcare and clinical research projects.<br />

Kamal at present provides specialised<br />

market research, business & consulting<br />

services and India entry strategy services,<br />

including regulatory support services to the<br />

clinical research companies from North<br />

America. He is also an Honorary Advisor to<br />

the leading clinical research education and<br />

training companies in India.<br />

Email: info@cliniminds.com<br />

<strong>IPI</strong> 25


<strong>IPI</strong><br />

Patient recruitment and<br />

retention in clinical trials<br />

Patient recruitment and retention is the<br />

leading problem drug companies face in<br />

developing their new pipelines. As recruiting<br />

becomes more diverse and complex,<br />

pharmaceutical companies are striving to<br />

discover new innovative ways to facilitate<br />

recruitment and keep patients enrolled in the<br />

clinical trial. With more and more clinical<br />

trials being done on a global level, it is<br />

imperative to have an understanding of both<br />

government regulations and social protocol<br />

that accompany these new markets.<br />

Patient recruitment and retention remains<br />

one of the most significant challenges faced<br />

by the pharmaceutical industry today, and<br />

accounts for hundreds of millions of Euros<br />

being spent due to delays in the clinical<br />

development process. Speeding up clinical<br />

trials is one opportunity to cut costs, improve<br />

efficiency and bring new drugs to market<br />

faster.<br />

Operational delays in clinical trials reduce<br />

patent exclusivity time and shorten the most<br />

commercially productive phase of a<br />

product’s life cycle. The primary source of<br />

such delays is the recruitment and retention<br />

of patients who fit the trials’ requirements.<br />

More and more drugs on trial have been<br />

designed to attack very specific biological<br />

targets. This necessitates the need to<br />

identify, recruit and retain patients who fit an<br />

increasingly narrow medical profile.<br />

Therefore, pharmaceutical companies and<br />

clinical research organisations are seeking<br />

new ways to optimise recruiting operations<br />

and gain a competitive edge. Patient<br />

Recruitment and Retention in Clinical Trials is<br />

a new report from Business Insights that<br />

provides a comprehensive review of patient<br />

recruiting strategies, focusing on successful<br />

approaches and emerging trends from<br />

across the globe. Use this report in patient<br />

and physician recruitment and to identify<br />

successful strategies to combat the clinical<br />

trials productivity crisis.<br />

The principles of successful patient<br />

enrolment involve the blending of sitefocused<br />

support strategies with a clear value<br />

proposition delivered with personal<br />

communication expertise. Building a<br />

relationship of understanding with the<br />

investigative site team and the participant is<br />

the ultimate goal.<br />

Recruitment Survey<br />

Site surveys indicate that CROs know less<br />

than sponsors about recruitment and<br />

retention. CROs have little internal expertise in<br />

recruitment strategy, budgets and timelines.<br />

The surveys also demonstrate that sites want<br />

specific tools: they do not want paid media<br />

campaigns or call centres, but rather they<br />

want at least an hour of recruitment training at<br />

the study launch meeting, and they want a<br />

point of contact to resolve their challenges<br />

with enrolment and retention.<br />

Subject Recruitment<br />

Subject recruitment is critical to the success<br />

of clinical research, both for scientific return<br />

and financial viability. Research studies that<br />

fail to meet recruitment goals provide<br />

minimal scientific return and may have a<br />

negative financial impact on the institution.<br />

There is substantial up-front institutional<br />

investment in the initiation of each of these<br />

protocols (institutional review board [IRB]<br />

review, contract negotiations,<br />

administration). Inadequate recruiting may<br />

increase study costs, delay time to<br />

completion, and possibly invalidate a trial<br />

due to insufficient study power. Thus, even<br />

modest improvements in the subject<br />

recruitment process may pay large<br />

dividends in accelerating the bench-tobedside<br />

cycle of new drug therapies or<br />

medical devices.<br />

Extramural Recruitment Efforts:<br />

• Providing clinicians with multi-language<br />

flyers that are easy to read and IRBapproved<br />

• Providing clinicians with brochures that<br />

are available on the internet and for office<br />

distribution<br />

• Providing sample letters to clinicians and<br />

introducing them to this trial<br />

• Providing educational opportunities to<br />

clinicians and nurses introducing them to<br />

this trial<br />

• Providing information to pathologists<br />

regarding this trial including how they may<br />

contact physicians with a potential study<br />

subject<br />

• Announcements in newsletters and<br />

websites available to participants<br />

• Presentations at participant meetings<br />

Intramural Recruitment Efforts:<br />

• Providing IRB-approved, multi-language<br />

flyers and brochures to the offices of PIs<br />

and associates for distribution<br />

• Providing laminated charts with<br />

inclusion/exclusion criteria for PIs,<br />

colleagues, study coordinators and<br />

nurses<br />

• Reviewing medical records in order to<br />

identify potential subjects<br />

• Encouraging primary physicians to inform<br />

potential subjects about the study<br />

• Encouraging PI to maintain open<br />

communication with associates in order to<br />

capture potential new subjects<br />

Steering Committee Efforts:<br />

• Collecting data on failed screens<br />

• Continual evaluation of barriers to<br />

recruitment<br />

The Principal Investigator’s Toolbox:<br />

1 Flyer for potential subjects<br />

2 Brochure for physicians to display in office<br />

3 Letter to referring investigators<br />

4 Letter to area physcian<br />

5 Laminated chart with inclusion/exclusion<br />

criteria<br />

6 Primer on subject recruitment<br />

7 Primer on consent process<br />

8 Primer on subject retention<br />

9 Subject handbook<br />

Primer on Subject Recruitment<br />

• Avoid approaching potential subjects until<br />

trust and rapport has been built<br />

26 <strong>IPI</strong><br />

www.ipimedia.com


<strong>IPI</strong><br />

• Appropriate introductions to a potential<br />

subject must be done if PI is not the<br />

primary physician This is necessary to<br />

build trust in the PI and to reassure the<br />

subject that the primary physician<br />

supports enrolment and study plan<br />

• Assure subject and family that continuity<br />

of care is not contingent upon<br />

participation<br />

• Fully explain compensation for travel,<br />

parking, meals, etc while attending study<br />

visits<br />

• Explain opportunity to receive some<br />

primary care under the auspices of the<br />

study, with potential of medical costs<br />

• Explain opportunity to receive closer<br />

medical attention and exposure to newer<br />

medications<br />

• Highlight opportunity to contribute to the<br />

advancement of medical knowledge<br />

Primer on Consent Process<br />

• Consent and assent should be available in<br />

primary language of the subject<br />

• Ideally the PI and study coordinator should<br />

be present<br />

• Allow enough time to speak with the<br />

subjects<br />

• Have both parents present if a minor is the<br />

subject<br />

• Go over all items in the consent and<br />

assent forms with as much explanation as<br />

possible<br />

• Allow time for questions in each section<br />

• Provide a copy of forms to take home for<br />

discussion and thought<br />

• Provide a realistic assessment of time,<br />

procedures, length of trial, benefits and risks<br />

involved in participating in the study<br />

• Emphasise that voluntary withdrawal from<br />

the trial at any time will not affect relationship<br />

with physician<br />

• Provide copies of signed forms to subject<br />

Extramural Centre Recruitment Efforts<br />

Include:<br />

• Announcements in journals, newsletters,<br />

websites<br />

• Presentations at medical meetings<br />

Steering Committee Centre Recruitment<br />

Efforts Include:<br />

• Reimbursement analysis with modification<br />

as needed and supported<br />

• Training support<br />

Clinical Cores Centre Recruitment Efforts<br />

Include:<br />

• Training support<br />

• On-site support as needed<br />

• Help with IRB submission<br />

Developing and Implementing an Effective<br />

Recruitment Strategy<br />

• Improve patient motivation, creating and<br />

communicating the study’s value<br />

proposition to patients<br />

• Providing positive reinforcement at every<br />

visit<br />

• Acquiring and utilising educational/<br />

support study materials for patients and<br />

their families<br />

• Comparing incentives, what patients say<br />

they need and what an ethics committee<br />

will allow<br />

• Improving listening skills for site staff<br />

The Impact of Protocol Design on Patient<br />

Recruitment<br />

• Designing with the patient in mind<br />

• The quantitative and qualitative data<br />

required to accelerate patient recruitment<br />

• Analysis of clinical trial durations in<br />

accordance with therapeutic areas<br />

• Screening systems for recruitment<br />

• Chief investigator and responsibilities<br />

• Impact of changing inclusion and<br />

<strong>IPI</strong> 27


<strong>IPI</strong><br />

exclusion criteria<br />

• Extrapolating data from past trials to<br />

forecast future ones<br />

Current Changes in Regulatory Affairs<br />

• A legislative update in recruitment for<br />

clinical trials<br />

• Manufacturers’ and wholesale dealers’<br />

licences<br />

• Clinical trials authorisations<br />

• Surviving inspections of premises and<br />

procedures<br />

Patient Recruitment at Investigator Hub Sites<br />

• Identifying a hub site<br />

• Confronting the issues for pharma<br />

• Existing potential solutions<br />

• Cost issues<br />

• Hub sites advantages<br />

Patient Retention<br />

Recent reports have indicated that the<br />

Health Insurance Portability and<br />

Accountability Act (HIPAA) that came into<br />

effect in 2003 has adversely affected clinical<br />

trial conduct and patient retention by<br />

imposing significant time and cost burdens<br />

and by impeding clinical trial recruiting and<br />

patient retention, which in turn has deterred<br />

patient search efforts – locating patients lost<br />

to follow-up. This commentary will serve as<br />

the third of a series that will discuss<br />

strategies to enhance patient retention and<br />

finding patients lost to follow-up based on<br />

evolving interpretations of the HIPAA<br />

regulations.<br />

Challenges of Patient Retention<br />

When conducting a clinical study on a<br />

treatment or intervention, it is imperative to<br />

retain patients throughout the entire study<br />

duration and for a follow-up period, which<br />

can be months or even a decade for<br />

longitudinal studies. Patient retention for<br />

study duration and follow-up represents a<br />

major challenge and we must address this<br />

issue to avoid potentially devastating<br />

consequences to the clinical trial industry.<br />

In fact, estimates reported in 2003, before<br />

the impact of HIPAA, indicate that 85% of<br />

trials do not finish on schedule, 60% to 80%<br />

of clinical trials do not meet their<br />

chronological endpoints because of<br />

challenges in recruitment, and 30% of trial<br />

sites fail to recruit even a single participant.<br />

Additionally, disappointing statistics show<br />

that 26% of patients drop out after providing<br />

consent, and greater than 94% of studies<br />

are delayed due to failed enrolment / patient<br />

retention (including patients lost to followup).<br />

The situation is worsening in large part<br />

due to HIPAA regulations. In fact, from 1997<br />

to 2003, the percentage of studies<br />

completed on time decreased from 18% to<br />

6%.3,4 The costly consequences of trial<br />

delay are so staggering that each day of<br />

delay can equate to $220k, and a two-month<br />

delay can equate to $70 million potential<br />

product sales due to postponed launch.<br />

Strategies to Enhance Patient Retention<br />

We can enhance retention of participants<br />

by utilising appointment reminders, with the<br />

caveat that this process must be cognisant<br />

of participant privacy. Under HIPAA,<br />

postcard reminders are discouraged due to<br />

the possibility of a patient’s medical<br />

diagnosis being inferred by anyone seeing<br />

the postcard (by virtue of a study logo or<br />

clinic’s name). Therefore, if utilizing mail<br />

reminders, enclose in an envelope without a<br />

return address that includes the affiliated<br />

study group (e.g., the Diabetes Research<br />

Group). Additionally, a mail reminder should<br />

not resemble a medical bill as patients may<br />

delay opening.<br />

Utilising a mail reminder several days in<br />

advance, followed by a telephone reminder<br />

24 to 48 hours prior to a follow-up<br />

appointment is an effective strategy for<br />

patient retention. Utilising this method<br />

ensures that the patient’s protected health<br />

information (PHI) is not revealed.<br />

Reimbursement of patients is also a wellknown<br />

strategy for effective patient retention.<br />

However, the processes of reimbursement<br />

must be consistent with participant privacy.<br />

Patients can also be reimbursed with gift<br />

cards, books of stamps, and other monetary<br />

equivalents.<br />

Lastly, a critical element of patient<br />

retention strategies are plans that include<br />

patient search initiatives to locate those lost<br />

to follow-up quickly and efficiently, so they<br />

return to the study in a timely manner. It has<br />

been shown that if patients lost to follow-up<br />

are contacted expeditiously they will return<br />

to the study, either during the ongoing phase<br />

or for follow-up evaluations.<br />

Aiding patient retention through site<br />

selection and Management<br />

• Finding the best locations to suit your<br />

needs<br />

• Strategies to evaluate site and resource<br />

requirements<br />

• Accounting for patients’ needs and<br />

privileges in site selection<br />

• Medical monitoring and reporting of any<br />

serious adverse effects<br />

Retention of Patients in Clinical Trials<br />

• Creating and communicating the study’s<br />

value proposition to patients<br />

• Providing positive reinforcement at every<br />

visit<br />

• Acquiring and utilising<br />

educational/support study materials for<br />

patients and their families<br />

• Comparing incentives, what patients say<br />

they need and what an ethics committee<br />

will allow<br />

• Improving listening skills for site staff<br />

Key findings of the report<br />

Almost half of all trial delays are caused by<br />

patient recruitment problems. These delays<br />

cost makers of specialty products more than<br />

$500,000 in lost sales and they result in<br />

losses of over $8m for blockbusters.<br />

The internet is an important tool to recruit<br />

both patients and physicians, but remains<br />

under-utilised throughout the US, Europe<br />

and Japan.<br />

Clinical research continues to migrate from<br />

the US and Western Europe to cheaper<br />

offshore locations. Virtually all of the leading<br />

CROs and many pharmaceutical companies<br />

including Pfizer, Roche, GlaxoSmithKline<br />

and Novartis now have an established<br />

presence in India, China and Eastern<br />

Europe.<br />

In March 2007, the European Medicines<br />

Agency (EMEA) adopted a draft guideline on<br />

requirements for first in-man clinical trials for<br />

potential high-risk medicinal products, which<br />

could further tighten patient recruiting ■<br />

Dr. Rajam Jaishankar<br />

has conducted a<br />

number of updates,<br />

workshops with<br />

evidence-based<br />

medicine as the<br />

theme, in the field of<br />

reproductive medicine<br />

in all most metros in India. She is the founder<br />

member of the Tamilnadu Pharmaceutical<br />

Welfare Trust and the former member of the<br />

Infertility subcommittee of the Association of<br />

obstetrics and Gynaecology. She is<br />

associated with the “Indian Fertility Society”<br />

(IFS), the National Association of<br />

Reproductive and Child Health of India<br />

(NARCHI), the Federation of Obstetrician and<br />

Gynaecological Societies of India (FOGSI)<br />

and the European Society of Human<br />

Reproduction and Embryology (ESHRE).<br />

Email: admin@questlifesciences.com<br />

28 <strong>IPI</strong>


<strong>IPI</strong><br />

Medical Imaging and Electronic<br />

Data Capture in Clinical Trials:<br />

the Future Paradigm<br />

All clinical trial data are ultimately digital. The<br />

pathway to this digital database is not always<br />

as connected as one might expect in this<br />

technological age. The backbone of clinical<br />

trials is now Electronic Data Capture (EDC),<br />

and this is not yet synchronous with the other<br />

major aspects of data collection, such as<br />

medical imaging. With the need for more<br />

streamlined processes, this paper explores<br />

the combination of medical imaging with<br />

EDC and provides the future paradigm for<br />

the triallist in the digital age. This will not only<br />

ensure an earlier final database lock, which is<br />

a key milestone, but during the trial data<br />

quality control (QC), and specifically image<br />

data QC, and evaluation can be performed in<br />

a more contemporaneous fashion with<br />

immediate feedback to those managing the<br />

study and the local investigator site. The<br />

ultimate goal is to unify all the essential data<br />

in a standardised format to expedite<br />

submissions and to increase the quality of<br />

those submissions.<br />

Introduction<br />

The use of medical imaging in clinical trials<br />

has seen an exponential growth in the last<br />

decade due to increased use of technology<br />

and improved computing power (1). This use<br />

continues to grow, particularly in early stage<br />

development when the latest techniques can<br />

aid in the early go/no-go decisions in new<br />

pharmaceutical/biotech<br />

product<br />

development. Examples of new techniques<br />

which are starting to be more commonly used<br />

in Phase I and II are listed in Table 1. For<br />

completeness, the more common<br />

applications that are used in Phase III are<br />

listed in Table 2, although the techniques are<br />

not exclusive to the phases listed.<br />

The need for an imaging core lab (ICL)<br />

has been described elsewhere (2) and is<br />

now a standard part of the team of the<br />

vendors in clinical trials (3). The days of<br />

leaving the image interpretation and analysis<br />

to the local investigator site have long gone<br />

due to the loss of data, reduced precision<br />

and lack of standardisation. The local<br />

investigators’ radiological department’s<br />

work is optimised for image acquisition and<br />

image interpretation for patient<br />

management, but not for the safety and<br />

efficacy reads.<br />

Along with the increased use of medical<br />

imaging in clinical trials, electronic data<br />

capture (EDC) has progressed significantly,<br />

so that in 2008 more than 50% of all trials<br />

used some form of EDC (4). The obvious<br />

question is raised about how to integrate the<br />

medical imaging and EDC into one system<br />

or process. This challenge is being<br />

addressed on many fronts. Historically the<br />

two processes have been managed<br />

“The information<br />

delivered by AG<br />

Mednet and others is<br />

not yet integrated with<br />

EDC systems, therefore<br />

the investigator site<br />

can provide<br />

information via the<br />

EDC system to the<br />

core lab about the<br />

incoming images”<br />

independently, and the ICLs have been<br />

totally separate entities from the EDC<br />

companies. The reason for this is twofold: 1.<br />

medical imaging is memory-, storage-, and<br />

transport-heavy, with some image files<br />

exceeding 10MegaBytes (MB) in size, and 2.<br />

the requirements in the radiology<br />

department are such that they either do not<br />

have access to the EDC system or the<br />

technologists do not have the knowledge to<br />

transfer the images to a system that takes<br />

them outside their institutional firewall.<br />

Convergence of the two processes is now<br />

starting to take place, although complete<br />

integration is highly unlikely for the<br />

immediate future due to the challenges<br />

mentioned. However we can anticipate<br />

some immediate developments and<br />

envision the full integration paradigm with<br />

the corresponding advances and<br />

improvements.<br />

This paper will discuss the concept of<br />

maximising the value of combining EDC<br />

and medical imaging, the utilisation of the<br />

synchronicity and working with ICLs and an<br />

EDC vendor with the current technology. A<br />

brief exploration of the mid-term future<br />

potential will also be entertained.<br />

Medical Imaging and Electronic Data<br />

Capture<br />

In most hospitals or clinics, medical images<br />

are available in a digital format, and even<br />

plain X-rays are obtained using digital X-ray<br />

although there are still some centres<br />

supplying plain film X-rays. The more<br />

technically savvy investigational sites will<br />

have the ability to submit the images to the<br />

imaging core lab via new technology<br />

processes such as those developed by AG<br />

Mednet (5). Not only are the images being<br />

sent electronically, but the data transmission<br />

information is sent and the images are<br />

anonymised. While this does not seem<br />

unreasonable in the current environment,<br />

there are still logistical and technical issues<br />

with some investigator sites, even beyond<br />

the issue of those sites still using film.<br />

Furthermore, the initial cost of setting this<br />

service up will be more than remaining with<br />

the well tried and tested method of sending<br />

the images via courier.<br />

Interestingly, using overnight courier<br />

services versus an electronic method only<br />

gains a few hours in the time of receipt of the<br />

images at the ICL, since many sites send<br />

their images at the end of the business day,<br />

regardless of which methodology is used.<br />

Therefore for the direct electronic transfer<br />

method the images will arrive at the end of<br />

the ICL’s working day and so will start being<br />

processed on the following work day. If the<br />

images are sent by courier they are routinely<br />

delivered by 10 or 11 am the following<br />

30 <strong>IPI</strong><br />

www.ipimedia.com


<strong>IPI</strong><br />

morning to the ICL, providing a net increase<br />

of time of delivery of less than four hours<br />

compared to the direct digital delivery<br />

system. However, there are additional<br />

advantages and disadvantages that have to<br />

be carefully considered with both methods;<br />

these are summarised in Table 3.<br />

The information delivered by AG Mednet<br />

and others is not yet integrated with EDC<br />

systems, therefore the investigator site can<br />

provide information via the EDC system to<br />

the core lab about the incoming images.<br />

Also, many EDC systems are tied in with<br />

interactive voice response systems (IVRS). If<br />

these are set up correctly, the ICL can<br />

receive notification at patient screening or<br />

randomisation, alerting them to the fact that<br />

there are screening images inbound, and<br />

they can be ready for the required rapid turnaround<br />

for eligibility of this data.<br />

Medical imaging endpoints are used<br />

primarily for efficacy, but are being used<br />

more for safety evaluation, e.g. bone density<br />

assessment in trials, like the ATAC study for<br />

breast cancer (6) or isotretinoin in acne (7).<br />

Medical imaging is also used as part of the<br />

eligibility criteria. The tie into EDC therefore<br />

becomes more critical, and the need for the<br />

ICL team and the EC team to discuss the<br />

trial data flow is important. It will no longer<br />

be acceptable to have these two groups<br />

working independently, since with the<br />

optimal logic built into the EDC, the imaging<br />

will take on a bigger role. EDC will capture<br />

the results of the patient management and<br />

safety reporting with adverse events. The<br />

results of the safety reads and possibly the<br />

efficacy reads will need to be entered to<br />

provide the comprehensive picture. EDC<br />

programming is not just a matter of entering<br />

the questions from the CRF but building a<br />

complete logic sequence to the data<br />

capture. If this is provided as a combined<br />

team, the correct logic sequence can be<br />

applied, ensuring that the principal<br />

investigators (PI) and study site coordinators<br />

can be “steered” through the process to<br />

ensure there is no missing data.<br />

➜<br />

Table 1. Novel Imaging Techniques used in Phase I/II Studies<br />

Molecular imaging techniques:<br />

Positron Emission Tomography (PET)<br />

Novel PET tracers,<br />

Dynamic Contrast Enhanced Magnetic Resonance Imaging (DCE-MRI),<br />

Magnetic Resonance Spectroscopy (MRS),<br />

functional MRI (fMRI)<br />

Finite Element Analysis (FEA)<br />

Active Shape Modelling (ASM)<br />

Hip Structural Analysis<br />

Table 2. Imaging Techniques more Commonly Used in Phase III Studies.<br />

X-ray<br />

Plain film<br />

Digital X-ray (DXR)<br />

Angiography<br />

Computerised Tomography (CT)<br />

Magnetic Resonance Imaging (MRI)<br />

Ultrasound<br />

Echocardiography<br />

Doppler Ultrasound<br />

Intima Media Thickness (IMT)<br />

Dual Energy X-Ray Absorptiometry (DXA) or bone densitometry<br />

Single Photon Emission Computerised Tomography (SPECT)<br />

Fundus photography<br />

Optical Coherence Tomography (OCT) for retinal imaging<br />

Table 3. The advantages and disadvantages of courier v direct electronic<br />

image transfer<br />

* Some older techniques require a “black box” to be installed and linked<br />

into the PACS system. This has been found to be unworkable at the site.<br />

<strong>IPI</strong> 31


<strong>IPI</strong><br />

Furthermore the logic should prevent<br />

incorrect data being entered. The author<br />

has experienced this first-hand in a complex<br />

osteoporosis study where with some round<br />

table discussions, the logic and<br />

performance of the EDC was enhanced and<br />

reduced the data entry at the site, while<br />

capturing the same information.<br />

The blinded reads that are conducted by<br />

the ICLs are, in most instances the two<br />

central radiologists (or readers, since they<br />

can also be rheumatologists, cardiologists<br />

etc) reading the images in isolation of all the<br />

data, with a third to adjudicate any<br />

differences (8). However, in oncology the<br />

readers need to know if the patient has had<br />

a resection or radiation therapy, depending<br />

on the study. This information should be<br />

available in real time to them via the EDC<br />

system. Many oncology reads often go on<br />

to a so-called “global assessment”. This is<br />

the process where a central oncologist will<br />

review the images with one radiologist and a<br />

set of clinical data that has been determined<br />

a priori. This provides a trial overview of the<br />

data and ensures there is complete<br />

congruity of the images and clinical data.<br />

Obviously the use of the EDC system makes<br />

this process much more seamless and the<br />

global assessment can potentially be<br />

captured in the EDC system. Historically<br />

these kinds of global assessments have<br />

been rate-limiting to the end of the study: the<br />

clinical data had to come into the data<br />

management department of the sponsor or<br />

the contract research organisation (CRO)<br />

and, once cleaned, shipped to the ICL for<br />

integrating into the read system. Only then<br />

could the final global read be conducted,<br />

often weeks or months after the last patient,<br />

last visit (LPLV) time point. With an integrated<br />

EDC system this process can be moved up<br />

to occur within days of LPLV and not slow<br />

down the data integration and database lock.<br />

Another aspect that has been gaining<br />

ground is the development of Adaptive<br />

Clinical Trials. These can only work if EDC is<br />

being used. In some of the more complex<br />

versions even the imaging may change<br />

depending on the arm or the inclusion<br />

criteria. This requires a very close<br />

relationship between the EDC vendor and<br />

ICL in the setup stages to ensure everything<br />

is captured in the correct sequence.<br />

As more imaging is used in Phase II, the<br />

cost-effectiveness of using the right EDC<br />

vendor to work with the ICL becomes<br />

increasingly important. Pressure on the<br />

pharmaceutical industry to develop process<br />

improvements at all levels has dramatically<br />

increased. Phase II studies are probably the<br />

last area to feel the pressure to compress<br />

timelines. EDC has formerly been seen as<br />

this cumbersome technology that requires<br />

months of programming and therefore not<br />

suitable to the Phase I/II environment. This<br />

is no longer the case and within an eight<br />

week lead time, EDC and imaging can be<br />

set up in an expeditious manner to provide<br />

clean and rapid data with the right partners.<br />

The Future<br />

As we conceptualise the future, combining a<br />

robust EDC platform with even some of the<br />

basic medical imaging technologies that are<br />

now being deployed will allow the image<br />

management to be built into the EDC<br />

component. The images can then be<br />

reviewed by the ICL and can be sent to the<br />

blinded read all within the same software<br />

platform. All the reads are then captured in<br />

the same EDC software platform.<br />

As we look into the future not only does<br />

this EDC synchronicity provide an elegant<br />

one-stop shop solution but it provides image<br />

data handling in the same environment as<br />

the other electronic data. Furthermore with<br />

proper planning and programming, the<br />

monitors and sponsor can have all this<br />

information in dashboard format providing<br />

advanced monitoring tools.<br />

Conclusions<br />

In the next 36 months or so, we can<br />

anticipate that medical images will be<br />

transferred electronically to the core lab as<br />

a de facto standard, and the courier will be<br />

relegated to the smaller investigator sites,<br />

new sites, or those sending X-rays. We<br />

can anticipate that those EDC vendors<br />

who are forward-thinking and linked in with<br />

ICLs will exploit the relationship and<br />

further improve the logic and questions in<br />

the EDC process. There will be a much<br />

closer tie-in with the IVRS and notifications<br />

to the ICL will result in a more rapid followup<br />

and knowledge of the incoming<br />

images. This will further reduce the losses<br />

of images that occur in clinical trials.<br />

The ultimate goal is to unify all the<br />

essential data in a standardised format, to<br />

expedite submissions, and to increase the<br />

quality of those submissions. With the<br />

advent of the e-clinical space, over time,<br />

medical imaging will become a<br />

specialised extension of this rapidly<br />

growing technology, rather than a standalone<br />

facet ■<br />

Dr. Collin Miller SVP<br />

Medical Affairs at Bio-<br />

Imaging, heads up the<br />

scientific over-sight<br />

within the company &<br />

provides consulting on<br />

trials in the muscularskeletal<br />

arena. He has<br />

a gained a Fellowship of the ICR (UK) and<br />

has attained the UK recognition of a<br />

Chartered Scientist. Dr. Miller has written and<br />

co-authored over 40 scientific publications<br />

and is co-editor of the books “Clinical Trials in<br />

Osteoporosis,” and “Clinical Trials in<br />

Osteoarthritis and Rheumatoid Arthritis,”<br />

published by Springer Ltd.<br />

Email: admin@questlifesciences.com<br />

References<br />

1. Reiber H, van Kuijk C, Schwarz, L.<br />

Medical Imaging and its use in clinical<br />

trials. European Pharmaceutical<br />

Contractor Autumn 2005.p80-84<br />

2. Miller CG, Noever K. “Taking care of your<br />

subject’s image: The role of Medical<br />

Imaging Core Laboratories” Good Clinical<br />

Practice Journal 2003, 10 (9) p 21-24<br />

3. Miller CG. Medical Imaging Core<br />

Laboratories. Applied Clinical Trials<br />

October 2005<br />

4. April 2007, Health Industry Insights<br />

#HI206351<br />

5. www.bioimaging.com/overview/agmednet<br />

6. Eastell R, Adams JE, Coleman RE et al.<br />

Effect of anastrozole on bone mineral<br />

density : 5 year results from the anastrzole,<br />

tamoxifen, alone or in combination trila<br />

18233230. J Clin Oncol. 2008, Mar<br />

1;26(7) 1051-7<br />

7. DiGiovanna JJ, Langman CB, Tschen EH<br />

et al. Effect of a single course of<br />

isotretinoin therapy on bone mineral<br />

density in adolescent patients with severe,<br />

recalcitrant, nodular acne. Journal<br />

American Academy of Dermatology 2004,<br />

Volume 51, No 5. p709-717<br />

8. Miller CG, Noever K. Reading Medical<br />

Images in oncology clinical trials.<br />

European Pharmaceutical Contractor 2004<br />

Summer, p 95-100<br />

32 <strong>IPI</strong>


<strong>IPI</strong> 33


<strong>IPI</strong><br />

Providing regulatory submissions<br />

in electronic format – US and<br />

EMEA actions<br />

The United States Food and Drug<br />

Administration (FDA) is taking concrete<br />

steps to require the submission of all types<br />

of regulatory communication in electronic<br />

format. Regulatory authorities globally are<br />

all moving to require electronic submission<br />

of regulatory communication. The reasons<br />

are many but the public safety<br />

responsibilities of the regulators require<br />

significantly faster access to content than<br />

can be supported by paper format.<br />

The cost of preparing the content to be<br />

reviewable in electronic format forms the<br />

bulk of the time and human resources<br />

needed to prepare electronic submissions.<br />

Failing to prepare the electronic content in<br />

compliant format can result in the regulatory<br />

authority refusing to conduct the review until<br />

the lack of compliance is corrected – this<br />

could have a profound impact on a<br />

company’s viability if a competitor drug is<br />

approved while the first sponsor is<br />

correcting non-compliant content.<br />

Compliance of electronic documents can<br />

be easily and inexpensively achieved<br />

through the proper use of Microsoft Word to<br />

author documents, combined with the use of<br />

templates with electronic submission<br />

compliance features built into the template<br />

design.<br />

1 HISTORY OF ELECTRONIC<br />

SUBMISSION REQUIREMENTS<br />

1.1 US FDA<br />

• FDA Centers for Drug Evaluation and<br />

Research (CDER) and Biologic Evaluation<br />

and Research (CBER) have required<br />

submission of electronic case report<br />

tabulations (CRTs) and case report forms<br />

(CRFs) in electronic format for new drug<br />

and new biologic license applications<br />

(NDAs and BLAs) since 1999 , .<br />

• FDA CDER has required package inserts<br />

to be submitted in electronic Structured<br />

Product Labeling (SPL) format since<br />

October 1, 2005 .<br />

• FDA CDER has required electronic<br />

submissions to be presented in the<br />

electronic Common Technical Document<br />

(eCTD) format since January 1, 2008 .<br />

• FDA is requiring the content of the Drug<br />

Establishment and Drug Registration<br />

forms to be submitted in electronic format<br />

beginning June 1, 2009 .<br />

• FDA has published the proposed rule that<br />

retired previous electronic submission<br />

formats, and set the stage to require all<br />

submissions be in electronic format using<br />

the eCTD organisation structure and<br />

electronic navigation aids .<br />

1.2 EMEA<br />

The electronic submission juggernaut is<br />

not limited to the US market:<br />

• The European Medicines Evaluation<br />

Agency has announced a timeline which<br />

requires all agencies participating in the<br />

centralised procedure for marketing<br />

authorisation applications (MAA) to begin<br />

submitting in electronic format on January<br />

1, 2009 , and in the eCTD format on<br />

January 10, 2010.<br />

Finally, there is an international effort<br />

underway with involvement by regulatory<br />

authorities and industry to define the next<br />

generation of bi-directional communication<br />

of regulatory submission content in a<br />

messaging format called Regulated Product<br />

Submission that is intended to address<br />

electronic submission requirements for all<br />

products regulated by FDA – drugs,<br />

biologics, devices, veterinary medicine, food<br />

additives, and cosmetics.<br />

2 TECHNICAL REQUIREMENTS<br />

Since 1999, the fundamental building block<br />

of electronic submissions has been the<br />

Adobe Acrobat Portable Document Format<br />

(PDF). However, this is not ordinary run-ofthe-mill<br />

PDFs created by scanning or<br />

printing to Adobe PDF. All regulatory<br />

authorities who accept electronic<br />

submissions have very strict requirements<br />

for the format and electronic navigation aids<br />

within PDF files, Regulatory authorities<br />

globally have refused to review electronic<br />

submissions whose PDF files lacked<br />

required navigation aids.<br />

All of these requirements can be met by<br />

implementing MS Word templates and<br />

configuring PDF conversion to support<br />

these requirements. PDF conversion can be<br />

done via PDFMaker, a plug-in to the<br />

Microsoft Office suite that is automatically<br />

implemented when the Microsoft Office<br />

applications are installed first, and Adobe<br />

Acrobat Standard or Professional are<br />

installed second. These properties can also<br />

be assured by correct configuration of<br />

Acrobat Distiller. Further, companies with<br />

MasterControl electronic document<br />

management systems (EDMS) configured<br />

for life sciences applications frequently have<br />

a PDF rendition server or generator in the<br />

EDMS suite that can be configured to<br />

support the requirements that are identified<br />

below.<br />

FDA has published guidance that<br />

describes the requirements for PDF files for<br />

eCTD submissions, Portable Document<br />

Format Specifications, version 2.0, dated<br />

2008-0604,www.fda.gov/cder/<br />

regulatory/ersr/ PDF_specification_v2.pdf.<br />

Other regulatory authorities have published<br />

requirements that are virtually identical to<br />

the guidance supplied by FDA.<br />

2.1 Fundamental PDF Requirements<br />

One of the strongest directives from the<br />

regulatory authorities’ requirements is that<br />

PDF files conform to predefined page sizes,<br />

have adequate margins on all sides and<br />

have adequate font sizes for legibility.<br />

These translate into the following<br />

specifications:<br />

• US letter page size for the FDA<br />

• A4 page size for rest of the world<br />

• Margins of at least one inch on all four<br />

borders, with larger margins on the<br />

binding edge in the event that paper<br />

volumes will be required<br />

• Times New Roman or Arial font, with the<br />

body text in 12 point and tables no smaller<br />

than 10 point<br />

34 <strong>IPI</strong> www.ipimedia.com


<strong>IPI</strong><br />

2.2 PDF File Format and Display<br />

Requirements<br />

PDF files should be in File Format 1.4 and be<br />

set to open to Fast Web View. These<br />

properties can be configured in the<br />

PDFMaker or PDF rendition process, but<br />

can be added to PDF files after the fact by<br />

using the PDF Optimizer function of Acrobat<br />

Standard or Professional.<br />

PDF files should automatically open to<br />

the page orientation of legibility. If<br />

PDFMaker, Distiller or a rendition engine is<br />

used to convert from MS Office to PDF<br />

format, this property can be automatically<br />

applied to the individual pages. However, if<br />

the PDF is created from scanned images,<br />

this requires the pages to be<br />

individually inspected and the page<br />

rotation property adjusted for pages<br />

that display incorrectly.<br />

2.3 PDF Files Created by<br />

Scanning<br />

Scanning is to be avoided unless<br />

there is no other choice. Scan<br />

documents of sufficient quality should<br />

be made searchable via Optical<br />

Character Recognition (OCR), which<br />

has been validated for accuracy of the<br />

recognition (for example that 1 has not<br />

been translated as l or i, or 0 has not<br />

been translated as O or o).<br />

If scanning must be done, the FDA<br />

guidance provides specifications that<br />

include:<br />

• Recommended resolution for<br />

different types of content<br />

• Compression algorithms for black &<br />

white, grayscale and colour<br />

• Image colour matching to assure<br />

fidelity of the depiction of colour<br />

for screen and print output<br />

formats<br />

2.4 Navigation via Hyperlinks<br />

and Bookmarks<br />

One of the most critical requirements to<br />

facilitate review is that PDF files contain<br />

“Compliance of<br />

electronic documents<br />

can be easily and<br />

inexpensively achieved<br />

through the proper use<br />

of Microsoft Word to<br />

author documents,<br />

combined with the use<br />

of templates with<br />

electronic submission<br />

compliance features<br />

built into the template<br />

design”<br />

hyperlinks and bookmarks to aid review in<br />

navigating within a file and between files.<br />

Examples of hyperlinks include:<br />

• From table of contents, list of figures, list of<br />

tables, list of appendices, to the content of<br />

the document<br />

• To supporting annotations, related<br />

sections, references, appendices, tables,<br />

or figures not located on the same page<br />

as the narrative text<br />

Bookmarks should match the table of<br />

contents hierarchy and depth, so that the<br />

document can be navigated via either the<br />

table of contents or the bookmarks.<br />

All bookmarks and hyperlinks should<br />

have relative file paths; that is not refer to<br />

server names, drive letters, or any<br />

company-specific network<br />

components.<br />

All bookmarks and hyperlinks<br />

should have the magnification<br />

property of Inherit Zoom, so that the<br />

reviewer’s view preferences are<br />

preserved when navigating via<br />

bookmarks and hyperlinks.<br />

Hyperlinks should be designated via<br />

thick blue lines or by blue text. If<br />

documents are created from an<br />

intelligent source – such as the MS<br />

Office applications – the blue text<br />

property can be applied at the time the<br />

documents are authored, and the PDF<br />

conversion process carries this property<br />

to the PDF file. However, if documents<br />

are scanned, hyperlinks can be<br />

represented only by thin blue lines.<br />

2.5 Page Numbering<br />

Individual PDF files in an eCTD should<br />

be page numbered beginning at page<br />

one, so that the PDF file and the<br />

document page number are the same.<br />

2.6 Conclusions on Preparing<br />

Compliant PDF Files for eCTD<br />

All of the PDF file properties described in<br />

the FDA guidance document can be<br />

assured by:<br />

<strong>IPI</strong> 35


<strong>IPI</strong><br />

• Using MS Word templates designed for<br />

eCTD compliance<br />

• Styling the document using Heading 1<br />

through 9 and Captions<br />

• Using MS Word properly – building tables<br />

of contents, tables of figures and tables,<br />

inserting hyperlinks or cross-references<br />

when referring to related content<br />

• Configuring PDFMaker, Distiller or PDF<br />

rendering engines to create compliant<br />

output<br />

• Be aware of the recommended<br />

configurations of resolution and colour<br />

when scanning documents<br />

These requirements apply not just to internal<br />

authors and regulatory operations staff, but<br />

should be made part of standard<br />

deliverables from contract writers, contract<br />

research organisations, and other outside<br />

suppliers.<br />

Correct preparation of PDF files for eCTD<br />

submissions ensures that the review starts<br />

on time and enables the review to be<br />

conducted with the support of the electronic<br />

navigation aids that are so critical to the<br />

review experience.<br />

3 FDA ELECTRONIC SUBMISSION<br />

GATEWAY<br />

FDA has implemented the Electronic<br />

Submission Gateway (FDA ESG) to enable<br />

sponsors to send information electronically<br />

to the FDA. It has been implemented using<br />

a software application certified to comply<br />

with secure messaging standards . The FDA<br />

Centers accepting submission via the ESG<br />

are listed in Table 1, along with the<br />

submission types that may be submitted via<br />

the ESG.<br />

FDA has made usage of the ESG<br />

inexpensive, requiring only digital certificates<br />

at the cost of US$20 per user per year, and<br />

an easy-to-follow process for registering for<br />

a test account and performing three test<br />

transmissions in order to qualify for a<br />

production account.<br />

EMEA and other health authorities<br />

around the world who accept electronic<br />

submissions have not implemented a similar<br />

capability for electronic transmission of<br />

regulatory communication, except for<br />

adverse event reports in the European<br />

Union.<br />

4 CONCLUSION<br />

Great strides have been made by the<br />

regulatory authorities in the US and EMEA to<br />

prepare their technical infrastructures for<br />

eCTD. Further announcements and<br />

clarification of intent to mandate electronic<br />

submissions are expected in 2008, with<br />

regular updates planned for this journal ■<br />

Antoinette Azevedo founded e-Submissions<br />

Solutions.com (a California corporation) to<br />

advise all sizes of<br />

biotechnology and<br />

pharmaceutical<br />

companies on the use<br />

of technology to<br />

manage regulatory<br />

documents and<br />

publish electronic<br />

submissions. Her experience ranges from<br />

developing strategies for document<br />

management and electronic publishing, to<br />

implementation of fully-validated systems,<br />

and production of paper and electronic<br />

submissions accepted for review by<br />

regulatory authorities worldwide.<br />

Email: AAzevedo@e-Submissions<br />

Solutions.com<br />

References<br />

1 Guidance for Industry: Providing<br />

Regulatory Submission in Electronic<br />

Format—NDA, U.S. Department of Health<br />

and Human Services, Food and Drug<br />

Administration, Center for Drug Evaluation<br />

and Research (CDER), January 1999.<br />

2 Guidance for Industry: Providing<br />

Regulatory Submission in Electronic<br />

Format—General Considerations, U.S.<br />

Department of Health and Human<br />

Services, Food and Drug Administration,<br />

Center for Drug Evaluation and Research<br />

(CDER), Center for Biologic Evaluation and<br />

Research (CBER), January 1999.<br />

3 Guidance for Industry: Providing<br />

Regulatory Submissions in Electronic<br />

Format —Content of Labeling, Food and<br />

Drug Administration, Center for Drug<br />

Evaluation and Research (CDER), Center<br />

for Biologic Evaluation and Research<br />

(CBER), April 2005.<br />

4 Guidance for Industry: Providing<br />

Regulatory Submissions in Electronic<br />

Format —Human Pharmaceutical Product<br />

Applications and Related Submissions<br />

Using the eCTD Specifications, Center for<br />

Drug Evaluation and Research (CDER),<br />

Center for Biologic Evaluation and<br />

Research (CBER), April 2006.<br />

5 Guidance for Industry: Providing<br />

Regulatory Submissions in Electronic<br />

Format — Drug Establishment and Drug<br />

Listing, U.S. Department of Health and<br />

Human Services, Food and Drug<br />

Administration, Office of the Commissioner,<br />

July 2008.<br />

6 Guidance on Providing Regulatory<br />

Submissions in Electronic Format:<br />

Withdrawal of Guidances, [Docket Nos.<br />

1999D-0054, 2001D-0475, and 2003D-<br />

0364] (formerly Docket Nos. 99D-0054,<br />

01D-0475, and 03D-0364, respectively)<br />

U.S. Department of Health and Human<br />

Services, Food and Drug Administration,<br />

7 EMEA Implementation of Electronic-Only<br />

Submission and eCTD Submission:<br />

Practical Guidelines Relating to Non-eCTD<br />

Electronic Submissions, London, 1<br />

November 2008, EMEA/633919/2008 v1.0.<br />

8 EMEA Implementation of Electronic-Only<br />

Submission and Mandatory eCTD<br />

Submissions in the Centralised Procedure:<br />

Statement of Intent, London, December<br />

2008, EMEA/572459/2008.<br />

9 Regulated Product Submission, U.S.<br />

National Institutes of Health, National<br />

Cancer Institute, www.<br />

gforge.nci.nih.gov/plugins/wiki/index.php?<br />

Regulated%20Product%20Submission&id<br />

=234&type=g<br />

10 Portable Document Format<br />

Specifications, U.S. Department of Health<br />

and Human Services, Food and Drug<br />

Administration, Center for Drug Evaluation<br />

and Research (CDER), Center for Biologic<br />

Evaluation and Research (CBER), version<br />

2.0, 2008-06-04.<br />

11 EMEA Implementation of Electronic-Only<br />

Submission and eCTD Submission:<br />

Questions and Answers Relating to<br />

Practical and Technical Aspects of the<br />

Implementation, London, December<br />

2008, EMEA/596881/2007, v0.5.<br />

12 FDA Electronic Submission Gateway (ESG),<br />

http://www.fda.gov/esg/, FDA Industry<br />

Systems Website Staff, May 1, 2008.<br />

13 FDA Electronic Submissions Gateway,<br />

Submission Types Supported by the FDA<br />

ESG,www.fda.gov/esg/userguide/WebHel<br />

p/AS2_Routing_IDs.htm#figure7http://ww<br />

w.fda.gov/esg/, FDA Industry Systems<br />

Website Staff, May 1, 2008.<br />

14 FDA Electronic Submissions Gateway,<br />

Submission<br />

Statistics,<br />

http://www.fda.gov/esg/submission_stats<br />

.htm , FDA Industry Systems Website<br />

Staff, May 1, 2008.<br />

36 <strong>IPI</strong>


<strong>IPI</strong><br />

Managing your flow with LIMS<br />

The evolution of Laboratory Information<br />

Management Systems (LIMS) from the point<br />

of view of a global clinical trials organisation<br />

Providing the backbone of a clinical trials<br />

organisation, a Laboratory Information<br />

Management System (LIMS) impacts on all<br />

departments, machinery and personnel. In<br />

today’s market no clinical laboratory or<br />

CRO could operate without the support of<br />

a well integrated LIMS package. And as the<br />

technology gets even more sophisticated,<br />

so do the opportunities for the<br />

pharmaceutical industry.<br />

LIMS exist to track clinical trial collection<br />

kits, manage and track specimens, manage<br />

laboratory testing workflows and quality<br />

performance, create laboratory reports and<br />

manage study-specific databases.<br />

Exclusive to hospitals in the early 1980’s,<br />

LIMS were originally used to manage patient<br />

samples and data on site. The systems<br />

began to evolve and were identified by<br />

clinical trials organisations as an effective<br />

way of managing the processes in<br />

commercial laboratories dedicated to drug<br />

development studies. A number of IT<br />

providers recognised the business<br />

opportunities available and began to mould<br />

the existing systems to suit the needs of<br />

clients working throughout the<br />

pharmaceutical sectors.<br />

By the 1990s most clinical laboratories<br />

had installed some form of LIMS system and<br />

in 1998, the FDA put into place regulation 21<br />

CFR part 11 to keep a tight grip on quality<br />

throughout the trials process. The regulation<br />

governs the scope and application of<br />

electronic records and signatures within the<br />

industry, with reference to validation, audit<br />

trails, record retention and record copying.<br />

As the regulations changed, so did the<br />

users’ need for more functionality – mainly<br />

driven by the pharmaceutical companies.<br />

Within a clinical trials organisation,<br />

laboratory staff wanted the LIMS to<br />

incorporate quality control and trends<br />

emerging from the studies with significant<br />

changes from pre-study samples and tools<br />

were created to perform these tasks.<br />

Project managers also recognised the<br />

benefits of integration into the LIMS<br />

package. As client demands increased, the<br />

project management functionality developed<br />

to provide the capability to interrogate the<br />

38 <strong>IPI</strong><br />

www.ipimedia.com


<strong>IPI</strong><br />

data once it was captured. To this end, the<br />

search engines evolved, front end reports<br />

were added and the overall user interface<br />

began to change by developing monitoring<br />

and reporting systems.<br />

The globalisation of the clinical trials<br />

industry has played a big part in the<br />

development of LIMS. The increase in multisite<br />

studies has forced the need for a<br />

centralised database of patient and product<br />

data which is fully compliant, secure, and<br />

accessible.<br />

The LIMS reports need to be available to<br />

all relevant personnel, sometimes on a<br />

global scale, including all the internal<br />

clinicians and project managers and<br />

externally to the doctors, monitors and<br />

sponsors. This is usually delivered through a<br />

front end system which allows different users<br />

varying levels of access depending on the<br />

need. For example, data may be blinded to<br />

a doctor carrying out the dosing on a<br />

diabetes study where 50 per cent of the<br />

patients are on the placebo. More<br />

sophisticated LIMS should also provide you<br />

with the capability to deliver the report in a<br />

number of ways – from hard copies, single<br />

or cumulative reports to remote data access<br />

systems.<br />

One of the more recent developments in<br />

LIMS is the traceability of samples<br />

throughout the clinical trials process from<br />

despatch by the investigator, arrival at the<br />

lab, through the testing procedure, right to<br />

freezer storage and disposal. Client<br />

requirements have to be met for full sample<br />

auditable history reports for every sample<br />

received in the laboratory listing for example<br />

sample freeze/thaw cycles or storage<br />

temperatures.<br />

Whilst a LIMS database cancels out the<br />

need for manual data input, it doesn’t take<br />

away the need for trained personnel to<br />

manage the processes of distribution,<br />

sample storage and other manual laboratory<br />

work. However, it does provide you with the<br />

capability to process more samples,<br />

forecast the timings of studies, monitor<br />

patient safety and work more effectively<br />

throughout the lab procedures.<br />

The system also allows you to measure<br />

the output capabilities of the laboratory,<br />

project the workload and any backlog, and<br />

forecast the number of personnel needed to<br />

perform the work. The LIMS database will<br />

also record trends and help forecast the type<br />

of work in the months ahead, for example,<br />

seasonal studies on winter viral infections.<br />

Once the scope of the study has been set up<br />

on the LIMS, accurate projections of<br />

workload can be calculated, subject to<br />

timely patient recruitment!<br />

There are relatively few IT companies<br />

which offer LIMS packages across the UK<br />

and Europe. Those that do have often<br />

“advanced security, data<br />

integrity, and audit trail<br />

capabilities should<br />

meet the stringent<br />

requirements of<br />

pharmaceutical<br />

clients”<br />

worked closely with their clients over the<br />

past decade to evolve their system and<br />

deliver the ever increasing list of functions<br />

and benefits. Other clinical trials companies<br />

and CRO’s have chosen to develop their<br />

own systems on site, responding directly<br />

and exclusively to the requests made by<br />

their sponsors and in-house team. The<br />

majority of service providers have released<br />

multiple versions of their LIMS package,<br />

improved, updated and in line with any new<br />

regulations.<br />

For a clinical trials organisation looking for a<br />

good LIMS package, the essential<br />

requirements are as follows:<br />

• Compliance with regulations – an obvious<br />

one perhaps, but with the FDA regulations<br />

constantly evolving, a LIMS provider must<br />

be wholly up to date on the changes and<br />

additions to regulation 21 CFR Part 11;<br />

• Easy to use – the best LIMS packages are<br />

those that are web-based, offering<br />

flexibility to work from laptops at different<br />

workstations around the laboratory. On<br />

another point, your personnel are trained<br />

as scientists, not IT experts, therefore the<br />

time it takes to train the staff on the system<br />

should be minimal;<br />

• Flexibility – a package that can be easily<br />

adapted, both by the IT provider and the<br />

user, allows an organisation to respond to<br />

its clients needs more effectively and add<br />

functionality as the business grows;<br />

• Security – a system’s advanced security,<br />

data integrity, and audit trail capabilities<br />

should meet the stringent requirements of<br />

<strong>IPI</strong> 39


<strong>IPI</strong><br />

pharmaceutical clients. LIMS must also<br />

provide a robust back up system to<br />

support a laboratory’s disaster recovery<br />

strategy;<br />

• Compatibility – the system must integrate<br />

with a lab’s different analysers<br />

For a pharmaceutical company working with<br />

a clinical trial organisation, the most<br />

important elements of LIMS are as follows:<br />

• Compliance with regulations;<br />

• Flexibility – the capability to see your study<br />

in any format you require;<br />

• Timely – to have real-time results which<br />

allow you to review/amend the study as<br />

needed;<br />

• Integrity – for the LIMS to provide you with<br />

accurate and secure data which can be<br />

monitored and shared globally;<br />

• A critical monitor of patient safety data<br />

• Adaptability – the willingness of the clinical<br />

trials organisation to work with its LIMS<br />

provider to adapt the system the suit your<br />

requirements.<br />

So where can the technology take the<br />

industry in the future? It is likely to be a case<br />

of evolution, not revolution and<br />

developments will come in line with any<br />

amendment of the FDA regulations.<br />

As the technology improves, so will the<br />

functionality of LIMS and their ability to<br />

integrate with other systems. Already we’re<br />

seeing an advancement in doctors’ case<br />

reporting with the majority of them now using<br />

electronic CRFs. Clinical trials organisations<br />

are now required to send their patient report<br />

for inclusion in these CRFs, creating a much<br />

more conclusive reporting system.<br />

The improvements to LIMS will also be<br />

led by sponsors’ needs. As the end user,<br />

pharmaceutical companies are likely to<br />

request more sophisticated reporting<br />

systems which provide quicker and more<br />

accessible results particularly with regard to<br />

patient safety.<br />

In the future, interim reporting is likely to<br />

become more mainstream, giving the<br />

sponsor the opportunity to review the study<br />

“LIMS must also provide<br />

a robust back up<br />

system to support a<br />

laboratory’s disaster<br />

recovery strategy”<br />

at any stage in order to amend the dosage,<br />

recruit more patients or simply monitor<br />

progress. Another request raised by a<br />

number of sponsors is the ability to link the<br />

storage information to the end data<br />

including the freezer temperature and<br />

disposal requirements.<br />

With the ever increasing use of EDC<br />

(Electronic Capture Systems) within the<br />

clinical trials industry, all LIMS databases<br />

should be upgraded to provide functionality<br />

to directly communicate with these systems.<br />

Away from the laboratory, improvements<br />

will need to be made to the invoicing<br />

procedures using LIMS. Integration of<br />

financial information is already being<br />

developed to include project management<br />

fees, analytical costs, courier services,<br />

sample storage and electronic data transfer<br />

all of which require ongoing investment.<br />

As a business and management tool,<br />

LIMS is integral to any clinical trials<br />

organisation and the investment required is<br />

fairly minimal compared to the obvious<br />

benefits it provides.<br />

In summary, the efficient running of a<br />

commercial laboratory has become<br />

dependent on LIMS technology and, with an<br />

emphasis on responding to clients needs,<br />

the opportunities for the future are endless ■<br />

Sam Singh is Director of Information Services<br />

and a founder member of Pivotal<br />

Laboratories, now ACM-Pivotal. Sam has<br />

worked in central laboratories for nearly 15<br />

years and has considerable experience in<br />

handling laboratory data as it is being<br />

generated. He has spent several years within<br />

the healthcare sector, developing LIMS. He<br />

has extensive ‘hands-on’ experience in<br />

managing laboratory data within a central<br />

laboratory environment, including producing<br />

Data Management Reports, data cleaning<br />

and performing database structure and<br />

integrity checks. As a SAS programmer, as<br />

well as a MUMPS programmer, he has<br />

particular expertise in programming<br />

laboratory data formats for electronic transfer<br />

to a huge variety of sponsor database<br />

systems.<br />

Email: s.singh@acm-pivotal.uk.com<br />

40 <strong>IPI</strong>


<strong>IPI</strong><br />

E-freight – Solutions for tough times<br />

The SWISS approach towards saving<br />

the environment and manage cargo<br />

consignments in electronic form<br />

In the complex world of international air<br />

freight, where profitability is measured by<br />

minutes saved, kilograms shipped and<br />

litres of fuel burned, the Swiss penchant for<br />

precision and reliability is a valuable asset.<br />

For Swiss WorldCargo, it has indeed given<br />

the airline a steady foundation that has<br />

seen profits grow, even as the industry as a<br />

whole continues to tighten its belt.<br />

A new leap has been made in managing<br />

air cargo and unsurprisingly, IATA has<br />

chosen Swiss WorldCargo to pioneer this<br />

process. “No paper, thanks!” will be the<br />

motto of Switzerland’s quality cargo carrier<br />

from the beginning of next year, as it will<br />

record and handle cargo consignments in<br />

electronic form. The new approach is not<br />

only good news for the environment; it will<br />

simplify processes, enhance transport<br />

quality and save money, too.<br />

“We are honoured to be chosen by IATA<br />

to trial e-freight in a practical working<br />

environment and help this new facility<br />

achieve its worldwide breakthrough as<br />

swiftly as possible,” says Markus Loeffler,<br />

Senior Manager e-freight at Swiss<br />

WorldCargo. “We see our selection as a<br />

42 <strong>IPI</strong><br />

www.ipimedia.com


<strong>IPI</strong><br />

‘paperless pioneer’ by the airline sector’s<br />

umbrella organisation as a clear<br />

confirmation that Swiss WorldCargo is one<br />

of our industry’s most innovative cargo<br />

service providers. But we also view it as an<br />

incentive and a commitment to further refine<br />

this forward-looking cargo handling<br />

approach.”<br />

IATA’s overall aim is to ensure that air<br />

cargo consignments are handled<br />

electronically as extensively as possible by<br />

the end of 2010. This will save a vast amount<br />

of paper – an annual volume that would fill<br />

39 Boeing 747-400 freighters, the<br />

association has calculated. It should also<br />

save the partners in the logistics chain<br />

worldwide a total of USD 1.2 billion a year –<br />

provided all these partners are electronically<br />

linked up and are prepared to make the<br />

switch to the paperless handling approach.<br />

The new e-freight facility will initially be<br />

introduced at Zurich Airport – again on<br />

IATA’s initiative. In practical terms, this<br />

means that not only Swiss WorldCargo but<br />

all the parties involved in the local logistics<br />

chain will exchange their consignment<br />

details online. The corresponding portal will<br />

thus include links to Zurich’s forwarders,<br />

handling agents and distribution agencies,<br />

and to the Swiss customs authorities.<br />

SWISS doing well in tough times<br />

“2008 has been very challenging,” says<br />

Oliver Evans, Chief Cargo Officer for Swiss<br />

International Air Lines, who recently passed<br />

through Bangkok. “However, through<br />

“I think e-freight will<br />

snowball once it hits<br />

Asia, and then it can’t<br />

be stopped. All the<br />

knowledge and skills<br />

that we learn as we’re<br />

going along will<br />

accumulate, and will be<br />

available to whoever<br />

joins the initiative”<br />

various hedging strategies that were<br />

developed by Lufthansa Cargo (Swiss<br />

WorldCargo’s parent company), we<br />

managed to minimize the damage and<br />

focus on areas that remained profitable. We<br />

were also forced to introduce surcharges,<br />

which helped us recover some costs.<br />

Although not ideal, these strategies together<br />

allowed us to stay on course.”<br />

Indeed, despite capacity growing by 20%<br />

last year, SWISS reports a constant 84<br />

percent system-wide load factor. “To us, this<br />

is a proof that our strategy is robust, and<br />

something that we should continue to<br />

nurture.”<br />

While Swiss WorldCargo has managed to<br />

maintain its very high performance of<br />

previous years, its competitors have not.<br />

Swiss WorldCargo’s third Customer<br />

Satisfaction survey held in 2008, further<br />

emphasises that it has a competitive<br />

advantage against its business rivals. The<br />

objective of the study, which included 1702<br />

interviews in 9 languages and in 35<br />

countries, and 3 weeks of phone calls 24/7<br />

was to measure three levels of customer<br />

satisfaction:<br />

• Overall satisfaction and loyalty<br />

• Main service elements in the air cargo<br />

process chain<br />

• Sub-elements that gave additional<br />

information and insight to the key<br />

elements.<br />

Industry Evolution<br />

Starting at a time when Swiss International<br />

44 <strong>IPI</strong>


<strong>IPI</strong><br />

Air Lines was emerging from the ashes of<br />

Swissair, and with the industry still reeling<br />

from 9/11, Mr Evans has seen firsthand the<br />

small details which can pay off in spades<br />

further down the road.<br />

For instance, one of the areas which has<br />

refused to buckle to economic or industry<br />

pressure is special cargo, a relatively small<br />

but significant part of the total market.<br />

“Special cargo shipments grew quite a lot in<br />

2007, but this year has seen the overall trend<br />

slow a bit,” he says. “However, the very<br />

nature of specialty shipments is what keeps<br />

them profitable. The smaller a market is and<br />

the more acute the needs of our customers<br />

are, the more urgent the shipments become.<br />

Our track record in this specialized area has<br />

ensured that it plays a large role in the<br />

success of our company.”<br />

In addition to focusing on the fine details,<br />

Mr Evans is also heavily involved in the move<br />

to e-freight. “I think elimination of paperwork<br />

is one of the last frontiers of efficiency in the<br />

industry – sometimes we use over 20<br />

documents for one shipment, which is a<br />

practice ripe for irregularities and confusion.<br />

Not only can we save on the environmental<br />

and hard costs of using all that paper, but we<br />

can also save on the cost of irregularities<br />

rising from the fact that everyone has a piece<br />

of the information, but not the whole picture.”<br />

Due to their relatively small market share,<br />

Switzerland was not chosen for the first wave<br />

of pilots but was picked to be a member for<br />

the second stage. “We feel that the SWISS<br />

community is a very special one, with<br />

advanced technology and a tight-knit<br />

forwarding community, and we really felt that<br />

we belonged in the pilot program,” says Mr<br />

Evans. The company is in the final stages of<br />

testing, and expects to initiate live shipments<br />

before the IATA cargo symposium in<br />

Bangkok in 2009.<br />

Going Hi-Tech<br />

As proof of their commitment to high-tech<br />

industry solutions, in early 2008 SWISS<br />

became one of the first airlines to give up<br />

their 30-year old ‘legacy’ IT platform in<br />

favour of a sleek new system that was<br />

designed specifically for them. The move<br />

was the biggest investment the company<br />

had made since its inception in 2002.<br />

“Our old computer system did the job,<br />

but it only did the job, so we needed to<br />

upgrade. We finally settled on Mercator, the<br />

IT arm of Emirates Group, because they had<br />

a team in place that would customize the<br />

system for us and help get us up to speed,<br />

which was essential. Having a good product<br />

is only half the story – if you don’t know how<br />

to implement it, you still have a problem.”<br />

The biggest challenge that e-freight will<br />

face is industry-wide integration. “It must be<br />

an attractive global initiative,” says Mr<br />

Evans. “Before that can happen, every bank,<br />

“despite capacity<br />

growing by 20% last<br />

year, SWISS reports a<br />

constant 84 percent<br />

system-wide load<br />

factor”<br />

shipper, forwarder, handling company,<br />

airline, and customs authority must be ready.<br />

We need to bring some enthusiasm to the<br />

table so that after a few successful pilots,<br />

everyone will say ‘I want a piece of that!’”<br />

But, as mentioned previously, industry<br />

support won’t be enough if you can’t<br />

integrate the new system properly. Many in<br />

the Asian logistics industry are familiar with<br />

the ‘if it isn’t broke, don’t fix it’ mentality, but<br />

Mr Evans doesn’t think this will be a<br />

problem.<br />

“Thais are keen learners, as our team in<br />

Thailand had demonstrated by their love of<br />

their jobs. If you have a passion for<br />

something, you do it. I think e-freight will<br />

snowball once it hits Asia, and then it can’t<br />

be stopped. All the knowledge and skills that<br />

we learn as we’re going along will<br />

accumulate, and will be available to whoever<br />

joins the initiative. Of course, some people<br />

won’t be ready, and they’ll be left behind, but<br />

those who are prepared will reap the<br />

benefits.”<br />

Mr Evans thinks that Thailand will fare<br />

well with the inevitable change. “Thailand is<br />

one of the key Asian economies and offers a<br />

lot of promise for tomorrow, with good longterm<br />

business prospects. Our team here is<br />

very good and is driving growth in this<br />

market.<br />

Troubles Bring Opportunities<br />

“Looking into 2009, we’re cautious but<br />

optimistic. We believe we have the right size<br />

of fleet, and the right number of destinations.<br />

We will be replacing some of our A330-200s<br />

with A330-300s for more fuel efficiency, but<br />

besides that, we’ll keep doing what we’ve<br />

been doing.” Plans are also in place to<br />

increase frequencies into Bangkok and offer<br />

daily service, but these won’t be<br />

implemented until further down the road.<br />

“I’ve studied the facts and figures, but I<br />

am an optimist by nature,” says Mr Evans.<br />

“The world is globalizing, and you will<br />

inevitably see crises pop up from time to<br />

time. People become overconfident, make<br />

risky investments, and then a wobble<br />

becomes a crisis. But I believe the crisis<br />

we’re seeing now is healthy; it’s a painful<br />

correction of the overconfidence that got us<br />

here. At SWISS, we say that we need to be<br />

prepared to weather the storm, and the only<br />

thing you learn from history is that out of a<br />

storm come a lot of opportunities. You just<br />

have to be ready for them.”<br />

About Swiss WorldCargo<br />

Swiss WorldCargo is the airfreight division of<br />

Swiss International Air Lines Ltd. With a<br />

worldwide network serving more than 150<br />

destinations in more than 80 countries and a<br />

broad spectrum of services, Swiss<br />

WorldCargo earns genuine added-value for<br />

its customers and makes a substantial<br />

contribution to the earnings of SWISS ■<br />

Bernd Maresch is the<br />

Director Marketing<br />

Strategy & PR; at<br />

Swiss WorldCargo<br />

and Liaison Manager<br />

Lufthansa Cargo and<br />

partner. Having<br />

received his Master of<br />

Business Administration and Social Sciences<br />

from FriedrichAlexander University of<br />

Erlangen/Nuremberg, Bernd has acquired a<br />

wealth of experience within the Air Cargo<br />

industry. He started his career at Crossair AG<br />

and then climbed through the ladders at<br />

SwissCargo. His vision and foresight has<br />

enabled Swiss WorldCargo to reach the<br />

pinnacle within the Pharmaceutical and Life<br />

Sciences industry.<br />

Email: bernd.maresch@swiss.com<br />

<strong>IPI</strong> 45


<strong>IPI</strong><br />

Risk Management in<br />

Clinical Trial Shipping<br />

Every time clinical trial supplies or the<br />

samples which are generated during the<br />

lifetime of a trial are transported then an<br />

element of control is taken from your<br />

hands. In order to minimise the lack of<br />

control, this article will discuss what kind of<br />

risks shipments face when they are<br />

actually in transit and when they get to the<br />

site, so that an assessment can be made of<br />

those risks, and contingency planning can<br />

be used to reduce them as much as is<br />

feasible in a time- and cost-efficient<br />

manner.<br />

Contingency planning is an important<br />

procedure to protect your assets. The<br />

process begins with a risk assessment<br />

analysis and focus is on incidents which<br />

appear more probable and are likely to<br />

impact upon key elements of the transport<br />

chain. Risk factors vary by country and<br />

region, so this approach may be required to<br />

be done both globally and locally. From the<br />

evaluation of risk, likelihood and impact, you<br />

can consider what controls can be<br />

implemented and then define the roles and<br />

responsibilities in an incident response.<br />

Although not all incidents can be avoided or<br />

controlled, this approach to risk evaluation<br />

helps to protect patient safety, company<br />

liability, protocol validation, intellectual<br />

property and study timelines.<br />

What are the consequences if a<br />

consignment is lost or destroyed? First of all<br />

there is a delay in supply to the site. There<br />

may not be any replacement product. Some<br />

clinical trials material goes through years of<br />

manufacturing, and may go through 20<br />

different processes in order to get it to a<br />

point where it can actually be delivered to<br />

the site. Arranging a new supply may require<br />

taking excess material from another site, or<br />

from another study. If the study is for a<br />

seasonal disease, something like<br />

pneumonia or influenza, the start of the<br />

disease season may be missed, losing a<br />

potential 12 months. Patients are removed<br />

from trials, losing data, which means delays<br />

in licence application. That can have very<br />

serious financial implications, but may also<br />

affect the share price and reputation of the<br />

sponsor. If a subject is enrolled on a study<br />

with a big pharmaceutical company and the<br />

company then fails to deliver the drug to<br />

them, then what is the patient’s impression<br />

going to be?<br />

There are a number of commonalities<br />

which will be detrimental to shipments.<br />

Because of limited stability data on<br />

investigational product temperature<br />

excursions represent major problems.<br />

Mapping of routes assesses the average<br />

temperatures experienced over time, rather<br />

than assuming or hoping what is likely to<br />

happen. Whilst the climate and individual<br />

weather issues are things which no-one can<br />

“Traditionally shipments<br />

are made at the start of<br />

the week, so that they<br />

will be delivered before<br />

the weekend almost<br />

everywhere”<br />

control, choosing not to ship in extreme<br />

conditions can be prudent.<br />

When shipments must be sent, then<br />

using additional protection such as heated<br />

active containers or thermal blankets can<br />

lessen the likelihood of excursions. In the<br />

event of shipping extremely temperaturesensitive<br />

shipments, the thermal challenge<br />

can come from other cargo. Dry ice boxes in<br />

the same area of the hold, or stacked inside<br />

a Unit Load Device, may result in chilling of<br />

a carefully selected thermal box, and this<br />

should be mitigated by using good quality<br />

boxes and on occasion thermal wrapping.<br />

The internal configuration of the product<br />

can dramatically affect control of<br />

temperature. Tiny patient packs consisting<br />

of a very small vial with a lot of air around<br />

them make temperature control extremely<br />

difficult, due to a lack of thermal mass. It is<br />

possible to increase the amount of thermal<br />

48 <strong>IPI</strong><br />

www.ipimedia.com


<strong>IPI</strong> 49<br />

<strong>IPI</strong>


<strong>IPI</strong><br />

mass using phase change material, or water<br />

gel packs, in the product box with the<br />

material.<br />

Whatever stability data is available can<br />

assist in choosing appropriate packaging.<br />

If, for example, the worst thing that can<br />

possibly happen is that the material freezes,<br />

then a package should be selected that is<br />

less likely to freeze and more likely to keep<br />

the internal temperature at the high end of<br />

the temperature range.<br />

Once the consignment arrives in a far-off<br />

country, with airline employees who do not<br />

speak any English but can recognise the<br />

numbers 2-8 on the outside of a shipment,<br />

then the materials can be killed with<br />

kindness by being placed into the fridge.<br />

Part-thawed gel packs can bring the<br />

temperature down below freezing. Careful<br />

labelling is required to reduce the likelihood<br />

of this happening. There are pharmaceutical<br />

companies which no longer put the name of<br />

the company on the outside of their box just<br />

in case somebody sees a pharmaceutical<br />

company’s name and puts it straight in the<br />

fridge. It may be more appropriate that all<br />

the information about keeping it cold or<br />

keeping ambient temperature goes onto the<br />

airline paperwork instead, which should be<br />

arranged with your transport provider.<br />

Regulatory changes can have a<br />

substantial effect on shipment timelines. For<br />

example, last year unannounced changes to<br />

the export regulations in Russia interrupted<br />

supply chains for shipping patient samples<br />

for around two weeks. Having flexible<br />

options in place to hold shipments in frozen<br />

or refrigerated temperatures may be a<br />

worthwhile backup, but may be prohibitively<br />

expensive to have on hand just in case.<br />

Changes to regulations are not confined to<br />

Russia, or the Far East. Regularly changes<br />

are introduced with no notice at all by the<br />

USA and by South American countries. The<br />

lack of warning can result in shipments<br />

being held for weeks at a time whilst new<br />

import permits are applied for, or changes<br />

made to documents which may then need<br />

couriering to customs or to the regulatory<br />

authorities, during which time temperature<br />

will need to be maintained. A system for gel<br />

pack replenishment or for storing boxes in<br />

the airline fridge (once they are outside the<br />

validation period for the boxes if they contain<br />

frozen refrigerant) should be considered,<br />

and this may be offered by your transport<br />

provider.<br />

Interruption of air carrier handling is the<br />

one of the most common incidents. Air<br />

cargo disruptions can be caused by many<br />

different factors such as weather delays,<br />

equipment problems, regulatory changes, or<br />

civil unrest. Controls vary by region and by<br />

the available transportation infrastructure in<br />

“If the study is for a<br />

seasonal disease,<br />

something like<br />

pneumonia or<br />

influenza, the start of<br />

the disease season<br />

may be missed, losing<br />

a potential 12 months”<br />

a given country, but when such events occur,<br />

it may be useful to have arrangements in<br />

place to utilise other methods of transport<br />

including road and rail services. You need to<br />

choose a reputable courier or freight<br />

forwarder to take responsibility for your<br />

package.<br />

Communication is critical in responding<br />

to an event. Critical role players need to be<br />

informed when service interruptions and<br />

supply delays occur, and escalation policies<br />

and information conduits should be in place<br />

well before any event occurs. Being aware<br />

of regulatory problems, air traffic control<br />

strikes, inclement weather, public holidays,<br />

civil unrest, interruptions in critical supplies<br />

such as dry ice, or any incident that could<br />

disrupt shipment timelines, is vital.<br />

After all the work done to transport a<br />

shipment safely and within the necessary<br />

temperature parameters, there is a very<br />

significant possibility that the investigator<br />

site will take the consignment into their<br />

facility and then fail to store it correctly. This<br />

usually happens because the site personnel<br />

who have been trained are not there, for<br />

example because it is their day off or they<br />

are on holiday. Traditionally shipments are<br />

made at the start of the week, so that they<br />

will be delivered before the weekend almost<br />

everywhere. Many shippers are now<br />

calculating shipping date from when the site<br />

can take delivery, rather than forwards. If the<br />

site says “We want it on Thursday morning<br />

because that’s when the research nurse is<br />

in,” then the date is calculated backwards<br />

from that time, which may mean shipping it<br />

on Tuesday afternoon. Shipments may be<br />

sent on a Friday so that they are at the front<br />

of the queue for customs clearance on<br />

Monday morning in Jakarta, or Philadelphia,<br />

remaining inside the validation time for the<br />

shippers.<br />

Pre-empting potential problems at the<br />

site requires a very clearly agreed chain of<br />

custody. Training just the primary<br />

investigator on how to deal with the drug is<br />

rarely successful, because the majority of<br />

the time a research nurse, or the practice<br />

manager, will receive the material. Training<br />

multiple people will vastly improve the<br />

likelihood of a successful delivery with no<br />

temperature excursions.<br />

Whilst the process of packaging,<br />

shipping and sending clinical trials material<br />

may at first sight appear very simple, it is<br />

clear from this limited overview that there are<br />

numerous factors which might contribute to<br />

the failure of a consignment to reach the<br />

destination in ‘perfect health’ ■<br />

Sue Lee – World<br />

Courier (UK) Ltd. has<br />

a background in<br />

Microbiology and has<br />

worked for World<br />

Courier for 17 years.<br />

As Manager BioPharm<br />

Systems, Research<br />

and Development she oversees the logistics<br />

for multinational clinical trials. Sue provides<br />

consultation and technical expertise to<br />

shippers, sponsors, labs and World Courier<br />

working groups on training, BioPharm<br />

shipping, and dangerous goods.<br />

She is a qualified DGSA for road and rail and<br />

IATA trained for shipping Dangerous Goods<br />

including Radioactives, and is a member of<br />

the Institute of Freight Professionals (Grad.)<br />

Email: Sue.lee@worldcourier.co.uk<br />

50 <strong>IPI</strong>


<strong>IPI</strong><br />

Greater flexibility in pharma packaging<br />

by reducing lot sizes and increasing<br />

machine running times<br />

In numerous pharma companies, the<br />

following problem is being posed:<br />

shrinking lot sizes are constantly<br />

worsening the ratio of packaging<br />

system set-up time to running time. The<br />

contribution below brings into question<br />

the current customary 1:1 linking of a<br />

blister machine, inline printing system<br />

and cartoning machine and looks at the<br />

feasibility and possibilities of partially<br />

decoupled packaging processes.<br />

1. Introduction<br />

Falling lot sizes are increasingly presenting<br />

the pharma packaging industry with a<br />

challenge. The ratio of set-up time to<br />

running time is constantly worsening, with<br />

the result that in many cases line efficiency<br />

is below 30%. The conversion, cleaning and<br />

clearing process is being consistently<br />

tightened up, but the improvements which<br />

are achieved are not in themselves sufficient<br />

to solve the structural problem of shrinking<br />

lot sizes. With today’s standard 1:1<br />

combination in the pharma industry of a<br />

blister machine, an inline printing system<br />

and a cartoning machine, every partial<br />

conversion - not to mention every fault -<br />

shuts down the entire packaging line.<br />

The following analysis questions this 1:1<br />

combination and examines a variant<br />

involving partially decoupled processes, in<br />

which the blistering, cartoning and printing<br />

stages can be carried out independently<br />

of each other - possibly only on a<br />

temporary basis.<br />

The possibility of bundling together<br />

generally small packaging lots on the blister<br />

machine and finishing them in a<br />

subsequent, decoupled secondary<br />

packaging process is termed ‘Late Stage<br />

Customisation’ (LSC); this is a concept<br />

which has been developed by the Körber<br />

Medipak Group, with the aim of enabling<br />

machine running times to be increased<br />

despite shrinking lot sizes (figure 1).<br />

The BIB-BOB module is designed to be<br />

mobile, so it can be linked quickly and easily<br />

to different blister machines or cartoning<br />

systems. The blister can be printed with<br />

customer-specific or country-specific<br />

information either by a printing system on<br />

the blister machine, which can be<br />

configured quickly, or at a later time, as it<br />

returns from the bulk magazine.<br />

In order to determine the possible<br />

increase in efficiency from<br />

partially decoupled line combinations,<br />

MediSeal, together with the Institut<br />

für Konstruktionstechnik und<br />

Anlagengestaltung (IKA) in Dresden, has<br />

developed a simulation program which<br />

allows the throughput time and the line<br />

efficiency of a typical European pharma<br />

packaging operation to be mapped. The<br />

program allows a direct comparison of the<br />

efficiency of “classic” line configurations<br />

and partially decoupled packaging<br />

processes.<br />

2. Simulation of a pharma company with<br />

different line configurations<br />

Fig. 1: Below<br />

The Körber Medipak Group’s concept for<br />

increasing machine running times<br />

despite falling lot sizes in pharma<br />

packaging.<br />

One essential feature for temporary<br />

storage of blisters is a logistics module -<br />

known as a BIB-BOB 1 (figure 2), which<br />

a) automatically removes blisters from a<br />

packaging line or thermoformer, stacks<br />

them in an orderly fashion in a<br />

magazine, and, b) at a later point in time<br />

feeds them back from the magazine into<br />

a cartoning system.<br />

Fig. 2: Right<br />

The BIB-BOB module - a logistics unit<br />

for removing or feeding blisters in bulk.<br />

52 <strong>IPI</strong><br />

www.ipimedia.com


<strong>IPI</strong><br />

In the example described, a pharma<br />

packaging company supplies a product to<br />

30 European countries. The packaging lot<br />

sizes vary between 200 and 60,000 trade<br />

packs; the language, the number of tablets<br />

per blister or the number of stacks per<br />

carton may vary from batch to batch.<br />

Dosaging of the active substance in the<br />

tablet must remain constant in this simple<br />

analysis. Depending on the type of<br />

conversion, 60 to 120 minutes were built in<br />

for the corresponding conversion and<br />

cleaning times. Downtime and idle time<br />

were included - depending on the line - on<br />

the basis of 5 to 12% of production running<br />

time.<br />

For planned production of 1.6 million<br />

blisters (approximately 455,000 trade<br />

packs), the pharma packaging company<br />

has two one-track lines of mid-range<br />

performance available - each equipped with<br />

a digital printing system. The nominal output<br />

of the thermoformer (e.g. a CP400), for two<br />

different cutter formats, is 180 and 320<br />

blisters per minute respectively; for the<br />

cartoner (e.g. a P3000), the figure is up to<br />

300 trade packs per minute.<br />

The two lines are identical and process<br />

300 packaging orders one after the other in<br />

the pre-specified production sequence.<br />

Under the conditions outlined, the<br />

packaging company with the two lines<br />

would need 161 hours in total to produce<br />

the 1.6 million blisters (approximately<br />

455,000 trade packs).<br />

The chosen line configuration is<br />

described below as “classic”, i.e. the blister<br />

machine, printer and cartoner are<br />

permanently connected to each other in a<br />

1:1 ratio (CLC ) 2 .<br />

3. Late Stage Customisation<br />

In order to (theoretically) process the prespecified<br />

production sequence again, the<br />

pharma packaging company makes the<br />

following changes to its plant:<br />

a) Line No. 2 is split up: the P3000<br />

cartoner is replaced by a smaller model<br />

(e.g. a P1600) with a nominal output of max.<br />

160 trade packs per minute. Both the CP400<br />

thermoformer and the P1600 are connected<br />

to a BIB-BOB system. The blisters produced<br />

on this CP400 are stacked using BIB mode<br />

in carton magazines (Line 2a); they are then<br />

automatically unloaded - at a later time - into<br />

the P1600 (Line 2b). If necessary, a blister<br />

printing system can be placed upstream of<br />

the P1600.<br />

Line No. 1 remains unchanged, i.e. the<br />

blisters produced are transferred directly to<br />

the P3000 cartoner.<br />

b) In contrast with the previously identical<br />

distribution of production orders over the<br />

two lines, lots for countries with a demand<br />

for larger numbers of items are now shifted<br />

to Line No. 1, whilst smaller lots (up to a few<br />

thousand trade packs) are moved onto sublines<br />

2a/b. A manual packaging station was<br />

also set up for the smallest lots (less than<br />

150 trade packs), as in this case even the<br />

quick-conversion P1600 (Line 2b) would not<br />

be economical.<br />

Fig. 3<br />

The two different line<br />

configurations, CLC<br />

(a) and LSC (b)<br />

compared: in the LSC<br />

configuration, line 2<br />

was split in to two<br />

sub-processes 2a<br />

and 2b, each with a<br />

BIB-BOB unit, and<br />

cartoning output was<br />

reduced. A manual<br />

packing station was<br />

also added for very<br />

small lots (Line 3).<br />

Fig. 4<br />

Cumulative production<br />

quantity of the two line<br />

configurations over time.<br />

<strong>IPI</strong> 53


<strong>IPI</strong><br />

4. Comparison of production output on<br />

the classic CLC configuration and the<br />

modified LSC line configuration<br />

4.1 Comparison of throughput times<br />

Figure 4 shows the cumulative blister<br />

production of the two line configurations<br />

(green=LSC; red=CLC). It is striking how<br />

the respective production outputs diverge<br />

right from the start and the LSC variant<br />

manages a continuously higher output. The<br />

specified production volume of 1.6 million<br />

blisters (approximately 455,000 trade packs)<br />

is reached after 9650 minutes (about 161<br />

hours) in the “classic” line configuration<br />

(CLC), whilst including the LSC variant cuts<br />

this time to 6650 minutes (about 111 hours).<br />

Throughput time can thus be reduced by<br />

about 31%.<br />

If both line configurations were run for the<br />

same time, e.g. 9650 minutes, the LSC<br />

variant would enable 700,000 more blisters -<br />

i.e. about 44% - to be produced.<br />

The reason why the differences are so<br />

considerable can be illustrated by a Gantt<br />

diagram analysis (fig. 5).<br />

4.2 Comparison of line utilisation<br />

sequences<br />

The simulation distinguishes between and<br />

documents the different line conditions<br />

using 4 categories: production (green),<br />

conversion time (pink), fault (red) and<br />

waiting time (blue).<br />

Figure 5 shows a comparative section of<br />

the product sequence for Line 1, once in<br />

classic mode (CLC, lower bar sequence)<br />

and once in LSC mode (upper bar<br />

sequence). It documents the production<br />

times and downtimes for each individual<br />

line. In the case of LSC mode, it is striking<br />

how Line 1 has significantly more and<br />

longer production times (green bar) than in<br />

CLC mode. The reason for this is that in LSC<br />

mode only the larger lots remain on Line 1,<br />

so the ratio of production time to conversion<br />

time is substantially better.<br />

Figure 6 documents the production<br />

times and downtimes of each individual line<br />

over the course of the entire production run.<br />

The two bars on the left represent the<br />

classic CLC concept; the following four bars<br />

show the line configuration modified using<br />

Fig. 5<br />

Gantt diagram:<br />

production<br />

sequences,<br />

Line 1.<br />

Fig. 6<br />

Documentation of<br />

the running and idle<br />

times of all lines, in<br />

the “classic” CLC<br />

configuration (L)<br />

and in “LSC mode”<br />

(R).<br />

the LSC approach. According to this, lines 1<br />

and 2 achieve 38.5 percent overall<br />

equipment effectiveness (O.E.E.) on<br />

average, whilst the same line in LSC mode<br />

achieves an O.E.E. of 64%. Merely removing<br />

small lots from line 1 enables an increase in<br />

use of capacity of 22% and 29% (25.5% on<br />

average) (fig. 6).<br />

Line 2a (CP400 with BIB-BOB) achieves<br />

a similarly good use of capacity; its<br />

production sequence is no longer<br />

interrupted by faults and size changes on<br />

the cartoner side (e.g. just changing the<br />

blister stack height).<br />

The manual packaging station was<br />

hardly used in the example, so “waiting<br />

time” is a predominant factor here.<br />

5. Summary<br />

Particularly when packaging small<br />

production lots, partially decoupling lines<br />

and linking the individual line segments with<br />

a logistics (BIB-BOB) module leads to a<br />

significantly better utilisation of capacity and<br />

distinctly shorter throughput times.<br />

Furthermore, as a result of decoupling, faults<br />

and conversion times only affect the part of<br />

the line involved. The BIB-BOB module has<br />

been designed so flexibly that if necessary it<br />

can dock with existing machinery ■<br />

Dr. Ralph Blum is<br />

Vice President Sales<br />

and Markting at<br />

MediSeal GmbH. In<br />

2003,he joined<br />

MediSeal, a company<br />

well known as a<br />

specialist in the field of<br />

blister packing and sachet packing<br />

machines as well as cartoning machines.<br />

Ralph Blum holds a Masters Degree in<br />

Physics and a PhD in Management<br />

Sciences. He joined MediSeal’s parent<br />

company, Körber AG, in Hamburg in 1999 as<br />

a member of the group's M&A Team.<br />

1.Abbreviation of “Blister into a Box - Blister out of<br />

a Box”. 2.Classic Line Configuration..<br />

54 <strong>IPI</strong>


<strong>IPI</strong><br />

Drug Counterfeits – Subject of<br />

an Experts’ Discussion at the<br />

European Parliament<br />

Dr. Jorgo Chatzimarkakis, Member of the<br />

European Parliament and Chairman of the<br />

Life Science Circle, invited experts and<br />

stakeholders to discuss the threats of drug<br />

counterfeits in Europe. The event, which<br />

took place at the European Parliament in<br />

Brussels on December 9th 2008,<br />

highlighted the increasing concerns about<br />

fake medicines penetrating the supply<br />

chain and the urgent need for measures to<br />

prevent a possibly catastrophic loss of<br />

confidence in medicines with all the<br />

negative consequences of health<br />

conditions on individuals and on public<br />

health.<br />

While the final report of CEBR 1 to the<br />

European Commission in 2002 still found<br />

that: “Public Health is monitored closely<br />

across the EU by the relevant drug<br />

administrations and instances of counterfeits<br />

are thought to be relatively rare”, the experts<br />

agreed that appropriate action was needed<br />

to maintain the high level of security and to<br />

prevent counterfeiters abusing the<br />

opportunities resulting from new global trade<br />

arrangements which may decrease<br />

traceability of pharmaceutical products and<br />

increase the chances for counterfeiting.<br />

Tony Björk, Vice-President of the PGEU,<br />

the Pharmaceutical Group of the European<br />

Union, the European association<br />

representing community pharmacists,<br />

suggested that a precautionary approach<br />

should be taken and that the pharmacist<br />

should act as the last line of defence against<br />

counterfeits. He referred to the benefits of<br />

the existing distribution system and said that<br />

mass serialisation with authentication at<br />

pharmacy level offered a way forward but<br />

added that issues like data control, work<br />

flow and cost of the system still needed to<br />

be addressed. Like most of the other experts<br />

he pointed out that the distribution of<br />

medicines via the internet was a cause of<br />

concern. “We cannot dis-invent the internet”<br />

he said, “but we must not actively<br />

encourage internet distribution unless the<br />

safety issues are resolved”, and he added<br />

that the right of the member states to limit<br />

internet sales of prescription medicines must<br />

be respected.<br />

Although often accused of being<br />

one of the possible sources of fake<br />

medicines, parallel trade can be an<br />

additional watchdog in the supply<br />

chain, emphasised Mr Edwin Kohl,<br />

the President of VAD, the German<br />

association of pharmaceutical parallel<br />

distributors. Since parallel<br />

importers need a<br />

manufacturing/labelling<br />

licence to market products, they<br />

are subject to the same strict rules<br />

as all manufacturing pharmaceutical<br />

companies within the EU, and using<br />

unreliable sources would put their own<br />

existence at stake. He stressed that imports<br />

“We cannot dis-invent<br />

the internet, but we<br />

must not actively<br />

encourage internet<br />

distribution unless the<br />

safety issues are<br />

resolved”<br />

from outside the EU were not parallel trade<br />

in the European terminology. Mr Kohl raised<br />

the question of how the counterfeit product<br />

could enter the wholesale market, and<br />

provided a solution that offers security<br />

against counterfeits. He suggested that the<br />

purchasing wholesaler should request<br />

information on the purchase from the selling<br />

wholesaler and the manufacturer of the<br />

drug, who would provide all data on the<br />

product excluding the original<br />

purchasing/selling price. Such a<br />

procedure would make sure that the<br />

purchaser obtained 100% certainty about<br />

the origin of the acquired product and would<br />

thereby be enabled to exclude fakes from<br />

the market.<br />

Monika Dereque-Pois, Director General<br />

of GIRP, the European Association of<br />

Pharmaceutical Full-Line Whole-Salers,<br />

underlined the importance of a well<br />

functioning mechanism of the supply chain<br />

on the basis of knowing one’s supplier and<br />

customer. GIRP and its members<br />

acknowledge the high responsibility they<br />

have to guarantee the safe and efficient<br />

supply of all medicines to all patients.<br />

Delivering some 100,000 medicines from<br />

more than 3500 manufacturers throughout<br />

the whole continent to more than 150,000<br />

pharmacies, they play a key role when it<br />

comes to patient safety and protection<br />

against counterfeits. Mrs Dereque-Pois sees<br />

the main point of concern in unregulated<br />

internet and mail order sale of medicines.<br />

Insufficient control of licences and the lack of<br />

Good Distribution Practice (GDP)<br />

➜<br />

56 <strong>IPI</strong><br />

www.ipimedia.com


<strong>IPI</strong><br />

compliance of non-full-line wholesalers, as<br />

well as slow communications about<br />

detected counterfeits aggravate the<br />

problem. Because of the complex web of<br />

actors without clearly defined<br />

responsibilities, a solid action plan needs to<br />

be taken forward, she said. GIRP published<br />

a position paper on the subject which can be<br />

downloaded from their website. Furthermore<br />

GIRP put together an “Anti-Counterfeit Task<br />

Force” of experts from European full-line<br />

wholesalers, launched the “Safe Supply<br />

Chain Coalition” and take part in EFPIA’s<br />

steering committee on “Coding and<br />

Identification of Pharmaceuticals”. In<br />

addition GIRP took part in EGA’s (European<br />

Generic medicines Association) integrity<br />

pact, an initiative to secure the supply chain<br />

and to fight counterfeits.<br />

Equally GIRP took part in the WHO<br />

IMPACT initiative as well as in the ad-hoc<br />

group on counterfeit medicines and the<br />

European Commission’s consultation<br />

process. Monika Dereque-Pois stressed the<br />

importance of collaborating with all supply<br />

chain partners. She made it clear at the<br />

same time that in addition to legislation,<br />

transparency and awareness, technological<br />

solutions were also needed. The<br />

wholesalers’ “Zero Tolerance Policy”<br />

towards counterfeits could only be part of an<br />

holistic approach towards patient safety,<br />

addressing supply chain integrity,<br />

collaboration and communication between<br />

all stakeholders and the verification of<br />

product throughout the supply chain. Since<br />

actually there is no common technology to<br />

automatically capture product information<br />

nor a homogenous coding system for<br />

product identification she urged for a<br />

machine-readable solution which could<br />

consist of either barcodes, two dimensional<br />

codes or electronic identification.<br />

Regardless of the technology, a seamless<br />

use of harmonised standards throughout the<br />

supply chain was a prerequisite in the fight<br />

against fake medicines. Another essential<br />

element in this fight is the clear<br />

accountability of all actors in the supply<br />

chain who need to be covered by valid<br />

licences. In this context she made reference<br />

to the high number of licences given out in<br />

some member states for pharmaceutical<br />

wholesalers, of which only a small<br />

proportion is actively being used (Germany:<br />

16 out of more than 4000; UK: 11 out of<br />

1700; Denmark: 3 out of more than 500).<br />

Mrs Dereque-Pois concluded that to<br />

successfully fight against counterfeit drugs<br />

required a clear definition of all actors and<br />

their responsibilities, harmonised licences<br />

and control mechanisms, centralised<br />

databases for the verification of the licences<br />

for all actors and adequate sanctions for<br />

non-compliance.<br />

Mr Klaus Gritschneder from the mail<br />

order pharmacy Europa Apotheek, located<br />

in Venlo, Netherlands, explained the<br />

difference between listed internet<br />

pharmacies and non-authorised web<br />

pharmacies. VIPPS, the Verified Internet<br />

Pharmacy Practice Sites (VIPPS)<br />

programme, developed in the spring of 1999<br />

by the National Association of Boards of<br />

Pharmacy in the US in response to public<br />

concerns over the safety of pharmacy<br />

practices on the internet could be a way<br />

forward also for Europe. The National<br />

Association of Boards of Pharmacy (NABP)<br />

was established in 1904 to assist state<br />

licensing boards in developing,<br />

implementing, and enforcing uniform<br />

“Regardless of the<br />

technology, a seamless<br />

use of harmonised<br />

standards throughout<br />

the supply chain was a<br />

prerequisite in the fight<br />

against fake<br />

medicines”<br />

standards to protect the public health.<br />

Pharmacy boards from fifty states, the<br />

District of Columbia, three US territories,<br />

nine Canadian provinces, and two Australian<br />

states make up the association<br />

membership. A coalition of state and federal<br />

regulatory associations, professional<br />

associations, and consumer advocacy<br />

groups provided their expertise in<br />

developing the criteria which VIPPSaccredited<br />

pharmacies follow. VIPPS<br />

pharmacy sites are identified by the VIPPS<br />

hyperlink seal displayed on their website. By<br />

clicking on the seal, a visitor is linked to the<br />

NABP VIPPS site where verified information<br />

about the pharmacy is maintained by NABP.<br />

The public is also welcome to access the<br />

VIPPS site at www.nabp.net to search for a<br />

VIPPS internet pharmacy which matches<br />

their needs. The use of such a “Verification<br />

Stamp” makes the difference evident<br />

between authorised and unauthorised webbased<br />

pharmacies.<br />

Mr Thomas Hoffman of the German<br />

Packaging Excellence Centre PEC in<br />

Stuttgart, Dr Peter Golz from VDMA, the<br />

German association of machine<br />

manufacturers, and Mr Richard Mertens<br />

from Uhlmann Visiotec explained the<br />

technical possibilities of applying a<br />

consistent coding system from the smallest<br />

sealable unit (such as an individual blister<br />

pocket) to the largest transportation unit like<br />

pallets, enabling the verification of the<br />

authenticity and origin of a product, even<br />

after removal from the original sales pack.<br />

The code, which is most likely to be a data<br />

matrix in combination with a unique serial<br />

number, can be applied by online printers in<br />

different printing technologies, and would<br />

allow track and trace of the drug throughout<br />

the distribution chain.<br />

Regardless of the technical solutions<br />

used, it seems to be clear that tight<br />

monitoring of the players in the<br />

pharmaceutical market, the close<br />

collaboration of all parties in the<br />

supply/distribution chain, tracking and<br />

tracing of the products and appropriate<br />

sanctions for non-compliance are required<br />

in order to maintain the safe supply of the<br />

market with genuine medicines ■<br />

Tassilo Korab holds MSc in healthcare<br />

Management. He started his career as an<br />

International Sales Manager, and has been in<br />

the packaging industry for more than 25<br />

years. Focusing on patient compliance,<br />

standards and regulations for child resistant<br />

packaging and the war against counterfeits.<br />

Co-founder of HCPC Europe, Tassilo Korab is<br />

the MD of TKM HandelsgmbH, Austria.<br />

Email: tassilo@korab.at<br />

58 <strong>IPI</strong>


<strong>IPI</strong><br />

Pharmaceutical piracy<br />

Tracking and Tracing<br />

and Anti-Counterfeiting<br />

It is estimated that in 2010, counterfeit drug<br />

sales will reach 75 billion US dollars<br />

globally, an increase of more than 90<br />

percent from 2005. This figure is highly<br />

alarming. But most worryingly, we must<br />

realise that this is no long-term prediction<br />

anymore. The pharmaceutical industry,<br />

politics, organisations and associations<br />

not only need to get plans going for an<br />

effective protection of medical products - a<br />

prompt implementation is also necessary<br />

as soon as possible.<br />

According to the International Medical<br />

Products Anti-Counterfeiting Taskforce<br />

(IMPACT) a medical product is counterfeit<br />

“when there is a false representation in<br />

relation to its identity, history or source. This<br />

applies to the product, its container or other<br />

packaging or labelling information.”<br />

Pharmaceutical counterfeits often cannot<br />

be recognised at first, and sometimes not<br />

even at second, sight – the counterfeit drug,<br />

its primary and secondary packaging are<br />

just too true to the original. Besides<br />

enormous losses for the pharmaceutical<br />

industry, the immediate danger for<br />

consumers is more and more prevalent. The<br />

forecast of global counterfeit drug sales<br />

reaching 75 billion US dollars next year asks<br />

for quick action; a demand that is not easily<br />

met. Pharmaceutical companies are well<br />

aware of the threat and many have already –<br />

more or less efficiently – put security<br />

measures into place to stop counterfeiting of<br />

their products. Also, political and industrial<br />

organisations, associations and institutions<br />

have reacted to the problem by initiating<br />

projects and publishing drafts and<br />

proposals for national or international<br />

guidelines or legislation changes. Yet, up to<br />

the present, no common solution has been<br />

found and the fight against counterfeiting<br />

still seems to be at its very beginning,<br />

different attempts and experiments often<br />

leading to no clear results.<br />

From RFID to SNI<br />

In the middle of 2006, FDA abandoned its<br />

original concept to combat counterfeits and<br />

ensure traceability with Radio Frequency<br />

Identification (RFID). Recently new<br />

developments have been introduced at FDA<br />

in order to establish standards for a unique<br />

standardised numerical identifier (SNI) for<br />

prescription drug packaging. The FDA<br />

Amendments Act of 27th September 2007<br />

(FDAAA) imposes March 2010 as the<br />

deadline for developing or adopting an SNI.<br />

In January 2009 draft guidance was issued<br />

by FDA, requesting comments and<br />

questions from the industry. In a first step,<br />

FDA has defined a package-level SNI that<br />

consists of the serialised National Drug<br />

Code (sNDC) and is composed of the<br />

National Drug Code combined with a unique<br />

“no common solution<br />

has been found and<br />

the fight against<br />

counterfeiting still<br />

seems to be at its<br />

very beginning,<br />

different attempts<br />

and experiments<br />

often leading to<br />

no clear results”<br />

8-digit serial number for each individual<br />

package. This guidance is considered as an<br />

“initial step to facilitating other measures for<br />

securing the drug supply chain” and is only<br />

to be viewed as a recommendation.<br />

Standards for track and trace, authentication<br />

and validation are not included, but will be<br />

dealt with in one of several announced<br />

guidances and regulations. Californian<br />

ePedigree, the last remnant of the ambitious<br />

FDA plans for the US-wide implementation<br />

of RFID, is an electronic record to trace every<br />

step taken by a retail package of<br />

prescription drugs throughout the supply<br />

chain. The initial proposal envisaged 2009<br />

as the deadline. This deadline was initially<br />

delayed until 2011, and now even until 2015,<br />

to give the involved parties more time to<br />

prepare for the implementation.<br />

European Commission on the move<br />

While the draft guidance of FDA does not yet<br />

include the topic of traceability, the<br />

European Commission is engaged in finding<br />

a method to protect pharmaceutical<br />

products against counterfeiting and at the<br />

same time assuring the traceability of these<br />

products. In March last year, the European<br />

Commission therefore launched a public<br />

consultation in preparation of a legal<br />

proposal to combat counterfeit medicines<br />

for human use. The Commission observes<br />

that “there is evidence that Member States<br />

are starting to consider taking unilateral<br />

action to address the problem”. For this<br />

reason, a consensus needs to be found for<br />

pack-specific tracing and mass serialisation.<br />

In its consultation, the EC specifies: “Mass<br />

serialisation is based on a code on the outer<br />

packaging which ‘individualises’ the pack.<br />

The code is read with a reading device.<br />

Currently, various tamper-proof technologies<br />

to implement such a concept are under<br />

discussion by the industry.” Datamatrix and<br />

2D barcoding are considered to be such<br />

technologies; other devices are not ruled<br />

out. One of the 123 responses from<br />

stakeholders to the European Commission's<br />

public consultation was issued by the<br />

European Association of Pharmaceutical<br />

Industries and Associations. EFPIA,<br />

amongst others, argues that “a risk-based<br />

approach may no longer be appropriate to<br />

apply these measures, as counterfeiters are<br />

now targeting a growing range of medicines<br />

and will simply move to target any<br />

weaknesses in the supply chain.” EFPIA is<br />

decidedly in favour of the proposed ban on<br />

repackaging because “it provides an open<br />

door for any illegitimate party to infiltrate the<br />

legitimate supply chain with potentially<br />

dangerous products”. One exception should<br />

be made to the ban on repackaging: “Any<br />

legislation will have to make provision to<br />

60 <strong>IPI</strong><br />

www.ipimedia.com


<strong>IPI</strong><br />

ensure that clinical trials and other research<br />

activities are clearly out of scope of any<br />

legislation arising out of the Commission’s<br />

initiative”.<br />

Initial first layer of security<br />

The UK's Medicines and Healthcare<br />

products Regulatory Agency (MHRA), as<br />

well as other respondents, raises concerns<br />

over a possible general repackaging ban at<br />

European level. Such a regulation would<br />

imply that importers could no longer comply<br />

with the requirements of supplying products<br />

placed on the UK market with an English<br />

leaflet and English labelling of the primary<br />

and secondary packaging. Many<br />

stakeholders emphasised the importance of<br />

cooperation with the American FDA, and of<br />

following a multi-layer approach, meaning a<br />

consideration of many different aspects and<br />

a bundle of methods. The majority of the<br />

respondents opined that it would be<br />

“premature, ineffective and even counterproductive”<br />

to decide on only one specific<br />

safety feature. Some respondents also<br />

stressed the necessity of sufficiently long<br />

implementation times and warned against<br />

an increase in bureaucracy. Changes in the<br />

legislation “should not lead to an overhaul of<br />

the existing legal system”. A stepwise<br />

approach seems to be most effective. One<br />

of the most important points in this<br />

discussion is that no government or<br />

international institution has yet found a<br />

mutual consent as far as the technology for<br />

counterfeiting and/or tracking & tracing is<br />

concerned. As required by many<br />

respondents to the European Commission's<br />

consultation, a choice of different security<br />

and traceability features should be possible.<br />

EFPIA brings it to the point when<br />

emphasising that “while tamper evident<br />

features and the use of authentication<br />

technologies present an initial first layer of<br />

security, it must be noted that these features<br />

can potentially be copied and alone do not<br />

constitute an absolute barrier to reduce<br />

counterfeits”. Traceability devices do not<br />

necessarily guarantee the authenticity of the<br />

traceable product. Although very<br />

sophisticated in their development, RFID<br />

tags and datamatrix codes are still far from<br />

being reliable security features. As for many<br />

anti-counterfeit features, some recent cases<br />

have repeatedly shown that these security<br />

measures can be copied almost as quickly<br />

as they enter the market for the protection of<br />

pharmaceuticals. What is more,<br />

counterfeiters are even ahead of the game.<br />

They apply security features to products<br />

that, in their original state, have no such<br />

feature. New implementations of anticounterfeiting<br />

devices are often not<br />

communicated to the public for fear of them<br />

being copied. Counterfeiters profit from this<br />

lack of communication.<br />

<strong>IPI</strong> 61


<strong>IPI</strong><br />

Vulnerable distribution chain<br />

The pharmaceutical supply chain may<br />

consist of twenty or more different steps<br />

before the drug reaches the patient. The<br />

production of each active pharmaceutical<br />

ingredient (API) is the first step of the<br />

distribution chain which has to combat<br />

counterfeiting. If not produced at the<br />

company’s own laboratory, each active<br />

ingredient will reach the production site<br />

through a network of suppliers. The different<br />

chemical substances are then combined to<br />

a pure drug substance to produce the final<br />

medicinal product. The European<br />

Commission confirms the risk of fake APIs:<br />

“Already the active pharmaceutical<br />

ingredients entering the manufacturing<br />

process may be false representations of the<br />

original API.” Therefore, choosing reliable<br />

manufacturers is, of course, an important<br />

prerequisite. The more people in contact<br />

with the drug, the greater the chance of an<br />

infiltration of counterfeits. The next step in<br />

the drug chain includes packing or filling.<br />

Additionally, secondary packaging and<br />

patient information leaflets are required<br />

before the medicinal product can finally be<br />

delivered to distributors or vendors. This<br />

rather simplified description of the<br />

pharmaceutical supply chain already reveals<br />

numerous steps which allow counterfeit<br />

drugs or packaging to enter the distribution<br />

chain, not forgetting the different routes<br />

travelled by the product to reach its final<br />

destination. Depending on the number of<br />

distributors and interim storages as well as<br />

on the number of countries or even<br />

continents, the product has to be<br />

dispatched by air, road or sea. Each mode<br />

of transport is exposed to the potential risk<br />

of counterfeiting. Pharmaceutical packaging<br />

is particularly prone to counterfeiting. The<br />

need for a technology that not only secures<br />

the primary and secondary packaging of<br />

medical products, but also ensures the<br />

authenticity of the traceability code and the<br />

information provided within is obvious.<br />

Colour-coded security<br />

One example for such combined technology<br />

is a system based on micro colour code<br />

technology to ensure a counterfeit-free legal<br />

supply chain. These micro colour codes with<br />

a size of 8 to 90 micrometers consist of four<br />

to eleven different colour-layers. The<br />

individual user code, which is exclusively<br />

allocated to and produced for the products<br />

of a specific brand-owner, is based on the<br />

combination of different colour-layers and<br />

their sorting order. A unique industry solution<br />

on the basis of these colour codes is now<br />

available for the combined tracing and<br />

securing of pharmaceutical products and<br />

constitutes an essential step towards<br />

improved security for the drug supply chain.<br />

The product, its primary or secondary<br />

packaging, its labels and seals can be kept<br />

secure by means of individualised colour<br />

codes. For example, secondary packaging<br />

is marked with a datamatrix code printed on<br />

a label to be used for tracking purposes. The<br />

colour code is also applied onto the label,<br />

thereby authenticating both the traceability<br />

code and the product. The same applies to<br />

primary packaging like tubes or blisters. In<br />

this example, the traceability code is now<br />

scanned with a 2D scanner and verified in<br />

worldwide existing databases. The product<br />

ID is checked and matched to the<br />

information provided in the database.<br />

However, even if the data and the<br />

information in the database coincide, the<br />

originality of the product is not yet<br />

guaranteed. If data verification reveals that<br />

the product has already been scanned, it is<br />

almost certainly a fake. Here, the authenticity<br />

of colour-coded products can immediately<br />

be verified by the use of a simple standard<br />

microscope. Manufacturers, distributors,<br />

police and customers are able to clearly<br />

distinguish between fake or original<br />

products and traceability codes. High costs<br />

of legal proceedings are avoided and<br />

customer trust in the company's products is<br />

maintained. The complete solution is<br />

obtainable from one single source and<br />

allows pharmaceutical companies to<br />

combine the logistic advantages of<br />

traceability with counterfeit-resistance and<br />

easy integration into the production process.<br />

2010 is approaching fast. And so is the<br />

predicted rise in counterfeited<br />

pharmaceutical products. American<br />

authorities revealed another alarming<br />

increase in fake pharmaceuticals: in 2008 a<br />

rise of more than 100 percent in comparison<br />

to 2007. The seizure of such high amounts<br />

of counterfeits is partly due to better<br />

collaboration among federal agencies. But it<br />

is mainly due to the overall increase in<br />

pharmaceutical counterfeits entering the<br />

legal supply chain. Much has already been<br />

done in order to find a solution to the<br />

growing problem. Yet even more still remains<br />

to be done. The different guidance drafts<br />

and public consultations are only the first<br />

step in a long discussion, decision and<br />

implementation process. Many different<br />

viewpoints and diverse conflicts of interest<br />

arise from this debate. No matter which<br />

codes or features pharmaceutical<br />

companies and national or international<br />

legislative bodies will opt for in the future,<br />

micro colour codes will contribute their share<br />

to the worldwide issue of tracking and<br />

tracing and anti-counterfeiting ■<br />

For further information, please visit<br />

www.3SGmbH.com.<br />

Nicole Golomb is<br />

Marketing and Sales<br />

Manager for anticounterfeit<br />

systems at<br />

3S Simons Security<br />

Systems GmbH,<br />

Germany. She is in<br />

charge of the management of the specific<br />

customer projects dealing with individual<br />

applications of the micro colour-code system<br />

SECUTAG® on branded articles. Additionally,<br />

Nicole is responsible for the analysis of<br />

processing technologies in specific industries<br />

and the product protection system's integration<br />

in the different sectors<br />

Email: nicole.golomb@secutag.com<br />

62 <strong>IPI</strong>


<strong>IPI</strong><br />

Subscription offer<br />

Ad index<br />

Guarantee you receive your copy of <strong>IPI</strong> –<br />

International Pharmaceutical Industry 4 issues<br />

per year. Pharma Publications is delighted to<br />

be able to offer its readers a great one years<br />

subscription offer.<br />

Subscribe now to receive your 20% discount<br />

POST: Complete the form detach and post to Pharma<br />

Publications, Diamond Key Building, Burwell Ind. Est.<br />

Burwell Road, London E10 7QG, United Kingdom<br />

FAX: Complete and fax this form to + 1 480 247 5316<br />

EMAIL: your details to info@pharmapubs.com<br />

Please tick the relevant boxes:<br />

UK & N. Ireland £80 inc p&p<br />

Europe 90 inc p&p<br />

USA $120 inc p&p<br />

I enclose a cheque made payable to Pharmapubs<br />

I wish to pay by credit card (Mastercard or Visa)<br />

Card number<br />

Valid from Expiry date Security code<br />

Name:<br />

Job Title:<br />

Company:<br />

Address:<br />

Post Code:<br />

Tel:<br />

Fax:<br />

Email:<br />

Signature:<br />

For Advertising oppertunities, contact Clive Baigent or Anthony<br />

Stewart on +44(0) 208 591 7584 Email: info@pharmapubs.com<br />

Page 11 ACHEMA 2009<br />

Page 19 Applikon Biotechnology<br />

IFC Bio Imaging Technologies<br />

Page 63 Bobst SA<br />

Page 20 Capsulution Pharma AG<br />

Page 59 Enestia Belgium NV<br />

Page 43 Envirotainer<br />

Page 41 Health Protection Agency<br />

OBC ICON Medical Imaging<br />

Page 9 INNOPHAR GmbH<br />

Page 5 ITI Lifesciences<br />

Page 15 Kendle International INC<br />

Page 55 Körber Medipak GmbH<br />

Page 22 MEK Consulting<br />

Page 24 Oxford Global – Drug<br />

Development Summit 2009<br />

Page 46 & 47 SCA Cool Logistics<br />

IBC Simons Security Systems<br />

Page 29 SMI – Accelerating Patient<br />

Recruitment & Retention in<br />

Clinical Trials<br />

Page 33 SMI – Imaging in Oncology<br />

Page 57 SMI – Pharmaceutical<br />

Packaging & Labelling<br />

Innovations<br />

Page 21 Strategic Diagnostics<br />

Incorporated<br />

Page 3 Swiss World Cargo<br />

Page 37 Thomson Reuters<br />

Page 51 World Courier (UK) Ltd.<br />

64 <strong>IPI</strong><br />

www.ipimedia.com


Excellence<br />

in Medical Imaging Solutions.<br />

Transforming images into information. Turning information<br />

into knowledge. Mapping life Pixel by Pixel. TM<br />

Experts in medical imaging for clinical trials. We provide phase I - IV integrated imaging solutions<br />

- from protocol design to regulatory submission - for pharmaceutical and biotech clients around<br />

the globe. With keen expertise and bold technology we transform images into knowledge.<br />

Our extensive range of experience includes expertise in:<br />

- Imaging Solutions for all Modalities & Therapeutic Areas<br />

- Clinical Trial Management Services for Diagnostic Imaging<br />

- Clinical Endpoint Committee Management<br />

T: +1 267-482-6300<br />

E: imaging@iconplc.com

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!