+ All Categories
Home > Documents > Copyright page ©2014 Saman Mazahreh ALL RIGHTS...

Copyright page ©2014 Saman Mazahreh ALL RIGHTS...

Date post: 05-Jun-2018
Category:
Upload: phamdung
View: 212 times
Download: 0 times
Share this document with a friend
164
Copyright page ©2014 Saman Mazahreh ALL RIGHTS RESERVED
Transcript

Copyright page

©2014

Saman Mazahreh

ALL RIGHTS RESERVED

THE DISARRAY IN THE REGULATORY APPROVAL PROCESS FOR

INTRODUCING NEW MEDICINES TO PATIENTS AROUND THE WORLD

By

SAMAN MAZAHREH

A Dissertation submitted to the

Graduate School-Newark

Rutgers, The State University of New Jersey

in partial fulfillment of the requirements

for the degree of

Doctor of Philosophy

Global Affairs

written under the direction of

Dr. Carlos Seiglie

and approved by

____________________

Dr. Carlos Seiglie

____________________

Dr. Douglas Coate

____________________

Dr. Mariana Spatareanu

____________________

Dr. Susan Sultzbaugh

Newark, New Jersey

May, 2014

ii

ABSTRACT

THE DISARRAY IN THE REGULATORY APPROVAL PROCESS FOR

INTRODUCING NEW MEDICINES TO PATIENTS AROUND THE WORLD

By: Saman Mazahreh

Dissertation Director:

Dr. Carlos Seiglie

The pharmaceutical regulatory framework is a complex system requiring great diligence

when requesting approval for a new medicine in different countries around the world.

The hypothesis was that cost of an application for a new drug approval would play a

major role in the country’s ability to review and approve a new drug. A study of the

history of regulation, current gaps in the system, and regional harmonization efforts was

conducted. Research and data collection on 129 countries was completed. Data included

economic, development, political, and health indicators. Statistical analysis was

conducted and some results confirmed part of the hypothesis while other results indicated

that further study is needed. Stability of a government was confirmed to be a key factor

in the drug approval process. The more stable the government, the more educated the

population, the higher life expectancy because of increased access to medicine.

As part of the research that led to the results above, it’s important to understand which

governance structure is in place or needs to be installed? What are the next steps moving

forward in gathering additional data from other companies, governments, and conducting

a bigger research project that can confirm the results of this study? The World Health

Organization, World Bank, International Monetary Fund, and other technical cooperative

groups serve as forums that are collaborating to achieve better faster results. The term

Global Affairs suggests that there are multiple subjects being discussed. The results of

iii

this paper imply and confirm that a multidisciplinary approach must be taken in order to

enhance the current global pharmaceutical regulatory process.

iv

Preface

The idea to venture into this work came about when I was working in Clinical Trial

Management for a major pharmaceutical company. The problem at the time was patients

in underdeveloped and developing countries did not have access to medicine that was

readily available in developed countries. At the time, it was the case of a drug I worked

on that is a potent chemical entity that fights infections. The drug received approval in

the United States and Europe and other developed countries were soon to follow. In my

role at the time, I was the project manager responsible for supporting all of the clinical

trials for that drug. Once approval was achieved, only a few clinical trials continued as

part of commitments to the health authorities. However, since not all countries had

approval or access to the drug, some patients in developing countries did not have the

option to receive that drug. We would receive letters from doctors in developing

countries indicating that their patients tried all of the current marketed medicines in their

country but were not cured. The doctors heard about our drug and wanted to get samples

for the patients. Unfortunately, it wasn’t as simple as sending a letter. In order to ship the

medicine, there were multiple regulatory hurdles, importation documentation, and proof

that the drug would not introduce an increased risk to the patient. In the end, we were

able to ship the medicine. In some cases, the patient’s health improved and we received

thank you letters; in other cases, the patient had ‘expired’ by the time the medicine

reached their hospital. So, today, we have this dissertation reviewing the current

regulatory process and some of the political and economic reasons of the current situation.

Access to medicine is getting better and global regulatory harmonization seems to be a

v

realistic goal. Below are some quotes that summarize the importance of access to

medicine.

From: FDA Global Engagement Report, April 2012

“As our world transforms and becomes increasingly globalized, we must come together

in new, unprecedented, even unexpected, ways to build a public health safety net for

consumers around the world.”

Margaret Hamburg, FDA Commissioner

“Today we recognize that to successfully protect U.S. public health, we must think, act,

and engage globally. Our interests must be broader than simply those within our own

borders.”

Margaret Hamburg, FDA Commissioner

“Globalization creates real opportunities to collaborate and leverage our collective

expertise and resources. Investments globally are critical to FDA’s success domestically.

Mary Lou Valdez, FDA’s Associate Commissioner for International Programs

“By helping countries build their regulatory capacities, we strengthen their power to

improve the safety and value of goods their own people consume, while also building

confidence in the imports they send to the United States.”

Margaret Hamburg, FDA Commissioner

From: George W. Merck’s famous “Medicine is for the people” speech made in 1950

https://www.merck.com/about/code_of_conduct.pdf

“We try never to forget that medicine is for the people. It is not for the profits. The

profits follow, and if we have remembered that, they have never failed to appear. How

can we bring the best of medicine to each and every person? We cannot rest until the

way has been found with our help to bring our finest achievements to everyone.”

Acknowledgement and Dedication:

I would like to acknowledge the unwavering support of Dr. Seiglie and my dissertation

committee members. Thank you! Without the committee members, I would not have

reached this point! Special thanks to Dr. Seiglie for his patience, leadership, and

continued encouragement that helped guide me through the challenges. Thank you for

caring. Special thanks to Dr. Coate for the inquisitive questions that made me think

differently about my approach to the problem. Special thanks to Dr. Spatareanu for

vi

actively supporting me, making me smile and having an open dialogue about industry and

economic theory. Last but not least, I cannot thank Dr. Sultzbaugh enough for her time

and dedication to this project while working full-time in industry and being a full-time

mom. Her great insight on industry, pharmacy background, and overall professionalism

has taken me to a new level.

I would also like to acknowledge Ann Martin and the Division of Global Affairs staff for

continuously raising the bar, and holding us to a higher standard. I would not be here

today without the continued support of all the staff. Thank you! I must mention and

thank my fellow PhD students for their continued support and creating a motivational

atmosphere to continuing working through all of the challenges, whether it is in the DGA

lounge, 8th

floor of Hill Hall, Library, or McGovern’s! I would like to thank all of my

friends and Kappa Xi Kappa Fraternity brothers for their support and encouraging me to

go over and above! I would like to thank my colleagues at work for their continued

support in guiding me through tough times or simply listening to my challenges.

Lastly, I want to acknowledge my family for their words of encouragement and believing

in me. I would like to thank my father for his words of wisdom in making the most

difficult issues so simple and for his words of empowerment and dedication. For setting

a great example of hard work paying off and to never give up. I would like to thank my

mother for her unending love, work ethic, and faith. I would not be the person I am today

if it were not for your teachings and installation of the belief to work for the greater good.

I would like to thank my siblings for their support, encouragement and always believing

in me. I would like to especially thank my nephews and nieces for always putting a

smile on my face and encouraging me to go over and above.

vii

Dedication:

I dedicate this to:

- My Grandmother, who has been calling me Dr. Saman since the day I was born.

While she was probably expecting and MD, I know she is watching me from

above with a smile on her face.

- My Parents for their unconditional love, teachings, inspirational conversations,

and helping me believe that I can do anything.

Saman Mazahreh

April 22, 2014

viii

Table of Contents

Abstract ii

Preface, Acknowledgement and Dedication iv

Table of Contents viii

List of Abbreviations and Acronyms ix

List of Tables xi

List of Illustrations and Figures xii

Introduction xiii

Chapter 1 1

Chapter 2 21

Chapter 3 49

Chapter 4 68

Chapter 5 86

Bibliography 92

Appendix I 96

Appendix II 101

Appendix III 126

Appendix IV 129

Appendix V 140

Appendix VI 142

Curriculum Vitae 148

ix

List of Abbreviations and Acronyms

AHC APEC Harmonization Center

AMRH The African Medicines Regulatory Harmonization

AP Asia Pacific

APEC Asia-Pacific Economic Cooperation

API Active Pharmaceutical Ingredient

ASEAN Association of South-East Asian Nations

CARICOM The Caribbean Community

CFR Code of Federal Register

CHMP Committee for Medicinal Products for Human Use

CIRS Center for Innovation in Regulatory Science

CMC Chemistry, Manufacturing, and Controls

CPMP Committee for Proprietary Medicinal Products

CPP Certificate of Pharmaceutical Product

CTA Clinical Trial Application

CTD Common Technical Document

DCP Decentralized Procedure

EAC East African Community

EC European Commission

EEMEA Eastern Europe, Middle East and Africa

EFI’A European Free Trade Association

EFPIA European Federation of Pharmaceutical Industries Associations

EFTA European Free Trade Agreement

EMA / EMEA* European Medicines Agency

EU European Union

FDA Food and Drug Administration

FD&C Food, Drug and Cosmetic Act

FIH First In Human

GCC Gulf Cooperation Council

GCC-DR Gulf Central Committee for Drug Registration

GCG Global Cooperation Groups

GCP Good Clinical Practice

GDP Gross Domestic Product

GMP Good Manufacturing Practice

HA Health Authority

ICDRA International Conference of Drug Regulatory Authorities

ICH International Conference on Harmonization

IFPMA International Federation of Pharmaceutical Manufacturers

Associations

IMF International Monetary Fund

IND Investigation New Drug

IOM Institute of Medicine

IRB Investigational Review Board

JP Japan; Japan Pharmacopeia

x

JPMA Japan Pharmaceutical Manufacturers Association

LSIF Life Sciences Innovation Forum

MERCOSUR Mercado Comun del Sur – Argentina, Brazil, Uruguay, Paraguay,

Venezuela, Bolivia

MoH Ministry of Health

MRA Mutual Recognition Agreement

MRP Mutual Recognition Procedure

NAFTA North American Free Trade Agreement

NCE New Chemical Entity

NDA New Drug Application

NGO Non-Governmental Organization

NME New Molecular Entity

OLS Ordinary Least Squares

PAHO Pan-American Health Organization

PANDRH Pan-American Network for Drug Regulatory Harmonization

PhRMA Pharmaceutical Research and Manufacturers of America

PMDA Pharmaceuticals and Medical Devices Agency, Japan

RHIs Regional Harmonization Initiatives

RMS Reference Member States

SADC South African Development Community

SC Steering Committee

SICA Central American Integration System

SwissMedic Switzerland Agency for Therapeutic Products

USA United States of America

USAID United States Agency for International Development

USP United States Pharmacopeia

WHO World Health Organization

*EMEA was the original name for the European Medicines Agency, but was then

amended to EMA.

xi

List of Tables

Table 1: Summary of laws and amendments introduced in the US regulations 43

Table 2: Variable Definitions 78

Table 3: Means and standard deviations of important variables 79

Table 4: Statistical analysis results using Ordinary Least Squares (OLS) model 80-82

Table 5: Statistical analysis results using Cox Hazard model 83

Table 6: Statistical analysis results using OLS model (using time instead of log.time) 143

xii

List of illustrations and figures

Figure1 – NDA review time per country (days) 5

Figure 2: NDA fees per country (USD) 6

Figure 3: NDA fee per country without the USA (USD) 7

Figure 4: Drug Development cycle for a new molecular entity (NME) 16

Figure 5: Standard NDA Regulatory Review Process and Milestones 20

Figure 6: Evolution of the ICH 55

xiii

Introduction

Theoretical modeling can help proactively identify faults or benefits of any system.

Theoretical statistical analysis can be very predictive and can help inform a system’s

potential output. Using empirical data that has been generated from actual outcomes in a

statistical analysis can yield powerful results that can confirm or dispute the theoretical

results. The approach taken in writing this dissertation combines all three pathways in

scoping the issue, leveraging industry data, and conducting statistical analysis to draw

conclusions.

The author has extensive experience in the pharmaceutical industry and therefore, a

practical approach was taken in the methodology and writing style. This approach and

writing style is slightly different than other dissertations that use the standard approach

that uses theoretical modeling in attempting to address an issue or answer a question.

The author used experience from the field, researched academic publications, collected

regulatory, economic, and political stability data in framing the situation. There are two

sides to the story (government and industry), and both have been represented in an

objective manner in this paper.

The diversity of pharmaceutical regulatory requirements makes marketing new drugs a

very complex and costly process that could delay public access to innovative and

essential drugs. In recent years, the pharmaceutical industry has become increasingly

global, taking advantage of the growing opportunities in the rapidly expanding new

markets in Asia, Latin America, the Middle East, and Africa. This globalization trend

created the need for a new strategic approach to pharmaceutical regulations, leading to

more international cooperation and harmonization. Globalization of pharmaceutical

xiv

regulatory standards has become a necessity and a goal for many groups of neighboring

countries in several regions of the world to reduce unnecessary and duplicative

requirement, rationalize time and costs, and create a transparent regulatory process that

improves access to medicines. Attempts to address this known gap have been fragmented.

In this paper, the following will be discussed:

1. Literature review of published work

2. History and current state of pharmaceutical regulation

3. Regional attempts to address the regulatory gaps

4. The role of politics and economics as it relates to the gaps

5. Draw conclusions, identify a potential path forward, and future research

Data has been collected form 129 countries1

on the lead-time for reviewing and

approving a New Drug Application (NDA) as well as the fees required by the respective

health authority. Economic data on the same countries was collected from the World

Bank; fragility score as well as democracy/stability index data were collected from the

Polity project for the same countries2.

The hypothesis is that countries with a higher GDP per capita, higher democracy score,

and lower fragility index will have a higher fee and shorter, predictive, lead-time for

review of new pharmaceutical drug applications. In other words, if the state is stable,

infrastructure will be in place to review the NDA in a timely manner for a set cost that the

applicant will be able to afford because the return on investment for the application will

yield substantial gains from sales.

1 See appendix I for a full list of countries surveyed and data collected

2 Data from the World Bank’s World Development Indicators were used in this study, see references

section.

In addition, data from the Polity project was used to better understand the political stability of each country.

See references section.

xv

The hypothesis will be addressed by first studying the evolution of the regulatory system

for the pharmaceutical industry. Then look at the regional harmonization efforts and how

countries have acknowledged that having separate systems for each country or even

region is not sustainable. Next, a statistical analysis will be completed of all the data that

has been gathered. The data set includes the results of the survey conducted, the data

researched from the World Health Organization, The World Bank, The Polity Index, The

Penn World Table, and other sources. The specific variables studied are:

- NDA review time

- NDA fees

- Life expectancy in a given country

- Literacy rate

- GDP per capita

- Health expenditure

- Physicians per 1000 people

- Democracy score

- Fragility index

- Openness to trade

A statistical regression will be run on the variables and results should show that a country

with high GDP, high literacy rate, and high health expenditure will have a high cost for a

NDA and short time for review of a NDA relative to the other countries.

Chapter 1: Literature Review

This section is broken into two parts. Part A focuses on the pharmaceutical regulation

sources and references while Part B identifies the references for the economic data and

regulation.

A. Pharmaceutical Regulation

Several sources3 have been used to generate the data and information contained in this

document. There have been several key websites, journals, an industry survey, and

textbooks researched to gain the amount of information needed for this dissertation.

Evidence from the available literature indicates there has been limited focus on the

discussion of global regulation and the economic factors contributing to the disarray in

global regulatory frameworks.

In looking at the overall spectrum of areas of research, a broad approach was taken to

retrieve information to support this dissertation. This approach is three fold: governance

structure, regional cooperation and local regulation. The first step is a review of the

current global governance structure that exists for all medicinal issues. The World Health

Organization (WHO), under the authority of the United Nations (UN) serves as the main

body that ties all of the public health issues globally4. Drug Regulation is one of the

topics covered by one of the committees within the WHO – International Conference of

Drug Regulatory Authorities (ICDRA). Previous conferences were researched and cited

for the content relevant to this research5. The conferences covered include: Hong Kong

3 Sources used include an industry survey, data from the World Bank, World Health Organization, and

ministry of health websites, among others. Refer to bibliography for a full list. 4 WHO is the directing and coordinating authority for health within the UN www.who.int

5 The ICDRA meetings occur every two years in different cities attended by Ministry of Health

representatives from most of the WHO’s member countries. The conferences cover multiple topics, one of

which is regulatory harmonization.

2

2002, Madrid 2004, Seoul 2006, Bern 2008, Singapore 2010, and Tallinn 2012. The

conferences occur every two years and representatives from the different ministries of

health discuss and develop plans to harmonize elements of the regulatory process. For

example, outcomes include decisions on the various governments adopting specific

protocols and specifications for approval of manufacturers of Active Pharmaceutical

Ingredients (API). However, no specific global legislation is proposed at these

conferences. Another organization that provides regulatory harmonization across all

continents is the International Conference on Harmonization (ICH). While the ICH does

not have authority to enforce guidelines or penalize organizations, the industry and

regulatory authorities have frequently adopted their published guidelines6.

Second, there are currently regional cooperation groups that discuss public health issues –

one of the main issues is regulatory harmonization. In this particular case, an example of

‘regional cooperation’ is the Gulf Cooperation Council (GCC) and Pan-American Health

Organization (PAHO). These organizations have developed milestones against certain

harmonization goals and review the plans during their regional meetings. Publications

from the conferences that were held were also reviewed and used for discussion in

Chapter 3 of this dissertation7.

Last, we have local (country level) regulatory bodies. These are usually under the

authority of the Ministry of Health (MoH). Where it exists, the country specific website

was researched for each country to confirm requirements for a new NDA. During the

research, it was observed that all of the websites focused on information for immediate

6 ICH has published guidelines in an effort to harmonize requirements. This will be discussed in more

detail in chapter 3. http://www.ich.org/ 7 Asia-Pacific Economic Cooperation. www.apec.org, APEC Harmonization Center. www.apec-ahc.org,

European Medicines Agency www.emea.eu , Gulf Cooperation Council. www.gcc-sg.org , Southern

African Development Community. www.sadc.int , Pan American Health Organization. www.paho.org

3

consumer need. It was very difficult to navigate through the websites and find the link to

the regulatory process. In many cases, no such link existed. The websites for the

countries are listed in Appendix II8.

In addition to reviewing the global governance structure for the health regulation, there

are also independent centers of excellence on regulatory requirements that have been

published on this topic. Information from their publications has been used for this work.

The Center for Innovation in Regulatory Science (CIRS) has evaluated specific country

requirements within specific regions to influence policy in the region9. Papers were

reviewed and elements of the study methodology were used to support the arguments

made in this paper. In a 201210

publication focusing on ICH countries (USA, EU, JP),

CIRS summarizes the NDA review timelines across the 3 major agencies (FDA, EMA,

PMDA)11

covering years from 2002 – 2011. The article highlights that there is a wide

variation in review times across agencies as well as within agencies primarily due to the

application category (priority vs. standard). Not all 3 agencies agree on which products

are priority versus standard. In the period between 2002 and 2011, 45% of the 249

approvals by the FDA and 17% of the 241 approvals by the PMDA were designated as

priority. This compares to just 5% of EMA for the same period of time (McAuslane and

Wang). McAuslane and Wang12

identified 69 common products that were approved by

all three health authorities from 2002 – 2011. Of the 69 products, 41 of the approvals in

8 Appendix II contains all of the websites found for each country that has a website. Some of the websites

are in the local language only. 9 Neil McAuslane and Tina Wang wrote the publication in March 2012 for CIRS titled: “New Drug

Approvals in ICH Countries: 2002 – 2011” http://www.cirsci.org/ 10

Ibid 11

The Food and Drug Administration (FDA) is the USA’s health authority. The European Medicines

Agency (EMA) is the European health authority. The Pharmaceuticals and Medical Devices Agency is the

health authority for Japan. 12

Ibid

4

the USA were priority reviews. McAuslane and Wang also highlight that the majority

(46) of the 69 applications were submitted to the FDA first, 26 of the 69 were submitted

to the FDA and EMA at the same time, while 53 of the 69 applications were submitted to

Japan’s PMDA more than a year after the initial submissions to the FDA and EMA13

.

McAuslane and Wang do not identify reasons for products being submitted

simultaneously to the US and EU regulatory authorities or why the Japan health authority

received most of the applications more than a year later. Japan requires local ethnic

clinical studies in addition to the basic applications submitted to the US and EU. These

studies are not usually conducted early in the drug development cycle because

manufacturers prefer to wait for the Phase III clinical study results to ensure a high

probability of success for the approval of the product, in any country. Since the

companies do not conduct the specific studies required for Japan only, the results of the

ethnic sensitivity studies come after the initial submission to the US and EU. As a result,

the new drug application to the Japanese health authority comes approximately a year

after the US and EU submission.

Another source of valuable data was industry survey that was conducted within a

pharmaceutical company. Industry data on approval time and fees were collected from

129 countries (plots of the data collected are included below). This survey was initiated

in 2011 and data collection was completed in 2012. Figure 1 shows the amount of review

time per country. The time is indicated in days and represents review of standard

applications, not priority or orphan drugs. The figure shows variability in many of the

countries where the most advanced countries are the troughs at approximately 365 days

and underdeveloped countries at the 3-year mark (1095 days). Due to the high number of

13

Ibid

5

countries, it was not possible to display the names of all 129 countries in Figure 1.

However, the data points for all the 129 countries are represented.

Figure 1 – NDA review time per country (days)

6

Figure 2: NDA fees per country (USD)

Figure 2 includes the entire list of countries. The US FDA price tag of $2MM skews the

table and most developing countries’ fees appear to be irrelevant in comparison. The

smaller peaks are the developed EU countries, Japan, Australia, and Canada. The

7

application fee was provided by most countries in USD. If the fee was provided by the

local currency, OANDA14

website was used to convert the cost to the USD.

Figure 3: NDA fee per country without the USA (USD)

When the USA fee is removed, the peaks in Figure 3 represent the European countries

with only Japan, Australia, and Canada with similar peaks. With this chart, countries like

14

Oanda is a currency converter website that is broadly used - http://www.oanda.com/currency/converter/

8

Singapore, Brazil, and Switzerland are the smaller peaks ranging from $50,000 to

$100,000 USD in application cost.

The Thomson Reuters IDRAC15

files have summaries of country specific requirements,

processes, and fees. However, the database is limited in the number of countries covered

and does not compare countries to identify gaps and opportunities. Information from this

source was used to compare data collected from a survey conducted with many of the

countries. See Appendix I for the regulatory processes from selected countries and an in-

depth description of the Singapore regulatory review process.

Additional academic publications on regulation were reviewed to better understand the

landscape of current published work. Braithwaite and Drahos16

discuss the regulatory

framework in multiple industries and focus on business regulation. A chapter of the book

dedicated to drug regulation focuses on the business as well as the current regulatory

regimes that govern the industry. The chapter begins by looking at the history of the

pharmaceutical regulatory process from its very inception and also discusses regulated

and unregulated drugs. Braithwaite and Drahos discuss key themes such as the EU

leadership in the harmonization of the technical requirements, the global regulatory

structure, regional, industry, individual, business, and professional actors in framing the

current landscape of the regulatory framework. Information from this text was used for

Chapters 2 and 3 as well as to build on the opportunities and challenges that currently

exist in the global regulatory process.

15

IDRAC files are available by subscription. A description of the available data can be found at the

following location: http://thomsonreuters.com/business-unit/science/pdf/IDRAC-cfs-en.pdf 16

Braithwaite, J., Drahos, P., Global Business Regulation, Cambridge 2000

9

Abraham and Smith17

compiled a collection of articles that discuss different points of

view in the pharmaceutical regulatory process. Relevant chapters included: The

regulatory laws and political culture in the United States and Germany, Europeanization

of medicines regulation, globalization of medicines control, New molecules, Markets and

changing drug regulatory practices, The limitations of current ethical regulations.

Abraham and Smith identify the gaps in the different regulatory authorities and stop short

of identifying a path forward.

Berry and Martin18

also contains a collection of articles covering a broad range of

regulatory questions mainly within the United States. The book also included topics on

generic drugs, biological, non-prescription (over-the counter medicine) and the impact on

the industry. These include: pharmaceutical regulation before and after the Food, Drug,

and Cosmetic Act, Modernizing the food and drug administration, The new drug

approval- process - before and after, Ways, means, and evolving trends in the U.S.

registration of drug products from foreign countries. This book focused on the US

regulatory process, and is supportive of the write up of the details outlined in Chapter 2 in

this paper.

In addition to published works, information from the ministry of health or drug regulatory

health authority websites for many of the countries was researched and used for

supportive information. See Appendix II for a listing of the countries with their matching

website. This is not an all-inclusive list and information extracted from these websites

gave additional details about the specific regulatory requirements in each country.

17

Abraham, J., and Helen Lawton Smith, Regulation of the Pharmaceutical Industry, New York : Palgrave

Macmillan, 2003 18

Berry, Ira, Martin, Robert P., The Pharmaceutical Regulatory Process, New York: Informa Healthcare,

2008.

10

Economic Regulation

The theory of economic regulation was studied as discussed by Peltzman and Stigler19

.

They argued that regulation is acquired by the industry and is designed and operated

primarily for its benefit. Alternative views of the regulation of industry include (1) the

notion that regulation is in place for the protection and benefit of the general public and

(2) the irrational political machine controls what it is directed to control but the few who

are in a position of influence. Stigler and Peltzman argue that the industry influences the

regulation to benefit itself only and this model is applicable across industries, including

the pharmaceutical industry. This theory may be applicable in the United States for most

industries, but this paper will show reasons why the economic theory of regulation does

not apply to pharmaceuticals, especially outside the US.

Ron Vogel, a Professor of Economics in the Eller College of Management and Research

Professor in the College of Pharmacy at the University of Arizona, covers a wide range of

topics on the pharmaceutical industry with focus on the economics and policy making

within the industry. Vogel argues that drugs were never regulated in the US until 1906,

when the Pure Food and Drugs Act was passed marking the beginning of the federal

regulation of drugs in the USA20

. Emphasis of the 1906 act was made on adulteration

and labeling of food, drinks and drugs, not testing and control of contents. In 1938,

Congress enacted the Food, Drug, and Cosmetic (FD&C) Act which required the drugs

19

George Stigler and Sam Peltzman published many articles on this top and a full listing is included in the

bibliography of this paper. Below are two of the articles referenced in this discussion.

Stigler, George “The Theory of Economic Regulation” The Bell Journal of Economics and Management

Science, Vol. 2, No.1 (Spring 1971), pp.3-21

Peltzman, Sam “George Stigler’s contribution to the Economic Analysis of Regulation” Journal of Political

Economy, 1993, vol. 101, no.5 pp.818 – 832 20

Vogel, Ronald, Pharmaceutical Economics and Public Policy, New York: Pharmaceutical Products Press,

2007

11

be proven to be safe for the use suggested on the label. FDA was given authority to

judge safety by requiring manufacturers to prove new drugs to be safe prior to marketing.

In addition, the law granted the FDA authority to regulate drugs and cosmetics in

addition to food. Therefore, companies started testing the drugs before marketing it to

comply with the laws. Amendments were introduced in 1962 to include more stringent

testing that increased the monetary burden on the pharmaceutical companies. Greater

safety thresholds were introduced mandating 3 phases of drug testing. For the first time,

companies were required to prove the drug was effective against the condition for which

it was being marketed for prior to marketing the product. Lastly, phase 3 studies, which

encompasses a larger patient population was added as a requirement to show both safety

and effectiveness of the drug. This regulation was a win-win situation for the regulators

and the patients21

.

Another valuable resource on the history of regulation and health economics is the World

Health Organization (WHO). In 2011 WHO published a report22

that provides details on

expenditures per country and per region. This information will be used in the

development of the argument for this paper in explaining rationale for identifying a

country with a firm infrastructure vs. those that do not. The 2011 report concludes that

per capita pharmaceutical expenditures in 2005/2006 ranged from $ 7.61 USD in low-

income countries to $ 431.6 USD in high-income countries, with considerable variation

between income groups in each country. The reports also highlights that the total

pharmaceutical expenditure is closely related with both total health expenditures, and

with gross domestic product (GDP).

21

Ibid 22

WHO Report WHO/EMP/MIE/2011.2.6

http://apps.who.int/medicinedocs/documents/s18767en/s18767en.pdf

12

The World Bank website is a valuable resource for providing details about the GDP and

GDP per capita for all the countries in this study. This information will be used to

support the argument that countries with stronger GDPs have a more developed

regulatory infrastructure to support regulation of medicines. The World Bank was also a

valuable resource in attaining information such as literacy rate, physicians per 1000

people, foreign direct investment (FDI), and life expectancy. While not all of the

information was directly used in the model, the information obtained provided supportive

background data in addressing the questions of this paper.

The Penn World Table23

was also a valuable resource for information on the countries’

financial status including real GDP from 1950 – 2011. This information will also be used

to support the argument that countries with stronger GDPSs have a more developed

regulatory infrastructure to support regulation of medicines. The historical trends from

this resource will also help show the progress being made in the emerging markets. The

primary data set used for this paper was the 2010 and 2011 GDP, the economy openness

factor (Open K) to show how much trade a country is involved in, and the size of the

government consumption (KG factor) relative to the GDP. This data set will be

discussed in more detail in Chapter 4. The Polity Project24

was a valuable resource in

attaining information on the democracy score and fragility index for the countries studied.

This information was used to confirm and support the argument of this thesis that the

more stable countries had a higher probability of having a sound pharmaceutical

regulatory process.

23

The Penn World Table can be found at : http://www.rug.nl/research/ggdc/data/penn-world-table 24

The Polity Index: http://www.systemicpeace.org/polity/polity4.htm

13

Overall, research was conducted in different disciplines using data collected from an

industry survey as well as leveraging published data. Combining these sets of data into

one paper and conducting analysis using statistical methods is a first. No other published

work has the combination of these data sets nor has any published work set out to test the

hypothesis set forth in this paper. Previously published work on regulation focused on a

single country, single region, or limited number of countries. In this paper, 129 countries

are included in the study. Other published work that focused on health related topics

discussed health expenditure and/or GDP, political stability, or regulatory process. No

published work combined these factors in a single publication. The following chapters

will discuss the history of regulation, the regional attempts at harmonization, the

economic factors as they impact the regulatory framework, and conclude with results,

potential path forward for the regulatory process, and future work.

25Prior to the submission of an NDA to a regulatory health authority, the manufacturer

must go through the drug development process. For the purpose of this paper, only a

brief description will be given to give background information on the content of the NDA.

A drug manufacturer must go through the initial toxicology stage to prove the new drug

being investigated is safe to test on humans. In order for this step to be complete, animal

studies are conducted where the animals (mice, dog, and/or monkey) are injected with a

strain of the disease being investigated and different dose levels of the new drug. Once

the company identifies the limits that would be appropriate to test on humans, an

Investigation New Drug (IND) Application is filed with the FDA (Clinical Trial

25

This section is written based on knowledge and experience the author gained from working in the

industry

14

Application – CTA in Europe26

). The application includes the toxicology data and a

proposed First-In-Human (FIH) study. The IND application is reviewed and, in some

cases, a special meeting is held with the regulatory authority in case of any questions.

The approval of the IND application marks the start of a Phase I study. Once the Phase I

study is complete, a Phase II study is planned, a clinical study protocol is drafted based

on the results of the Phase I study, and another application is submitted to the health

authority for approval prior to the initiation of the study. Once approval is granted, the

study can begin. If the study is to be conducted in more than one country, a separate

application must be filed to each individual country prior to the start of the study. Once

the Phase II study is complete and all results are analyzed, meeting is typically scheduled

with the regulatory authority to discuss results and plan for Phase III. A Phase III study

protocol is finalized; it is submitted to the health authority in the countries that will be

involved. Once approvals are received, the clinically labeled product is shipped out to

each country for study initiation. Phase III studies are typically global large-scale studies

that can take 2 -3 years to complete. Once the study is complete and results are reviewed

within the company and an expert panel, a decision is made to submit the NDA to the

health authority. The NDA contains information on all of the studies conducted starting

with initial discovery studies. The NDA also contains details on the chemical synthesis

of the new molecular entity and the drug product manufacturing process. The review

time shown earlier in Figure 1 reflects the review time for the NDA only, not the

previous phases.

26

A Clinical Trial Application is a form that a research organization submits to a health authority to request

permission to conduct a clinical study. More details can be found here:

http://ec.europa.eu/health/files/pharmacos/docs/doc2005/10_05/ca_14-2005_en.pdf

15

The other piece of important information is that, as the drug development cycle

progresses, the number of molecules being studied decreases. In the early phases of

discovery, thousands of compounds are investigated. Out of the thousands of compounds

studied, only a few hundred progress to the pre-clinical / toxicology / animal studies. Out

of the few hundred compounds, less than 10 are progressed to the Phase I FIH study. The

number of subjects tested in the Phase I study range from 20 to 100. Compounds

entering Phase II are between 5 to 10 and the number of subjects range from 100 to 500.

Fewer than 3 compounds from the original thousands reach the Pivotal Phase III stage of

development where massive global studies are conducted with thousands of patients from

around the world. The information from all of the studies conducted are included in the

NDA. Additional Phase IV (post approval) studies are conducted to monitor the market

activity. Some countries (other than the most developed) require 12-month post-approval

pharmacovigilance27

data. This is another reason some of the spikes are seen in Figure 1

shown earlier in this chapter. Figure 4 below summarizes the drug development process

for all of the stages.

27

Pharmacovigilance studies focus on the detection, assessment and prevention of adverse reactions to

drugs. In the context of this paper, these are studies that are conducted for drugs on the market.

http://apps.who.int/medicinedocs/en/d/Jh2934e/3.html

16

Figure 4: Drug Development cycle for a new molecular entity (NME)28

To simplify the description of the NDA review process, Figure 5 has been created to

show the generic drug approval process for many of the developing countries that require

a Certificate of Pharmaceutical Product (CPP)29

. A more detailed description of the

regulatory review procedure and process maps for selected countries can be found in

Appendix I. In most countries, a Registration Committee would be in charge of the

overall application and would be the primary contact with the applicant company. The

Registration Committee would have sub-committees that focus on the various sections of

the application. The first step in Figure 5 is for the health authority to validate or confirm

that the application is complete and contains all of the relevant sections. In some cases,

28

Graphic of the drug development cycle representing all clinical studies, number of patients in each phase,

as well as number of compounds studied: http://www.immunetrics.com/applications/drug-discovery.php 29

Certificate of Pharmaceutical Product (CPP) must be issued by one of the major countries (US, EU, JP)

manufacturing or marketing the product confirming the product meets all legal and quality requirements

and is being marketed in that country.

http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/ImportsandExportsCompliance/uc

m348825.htm

17

the health authority may request additional information from the company that is filing

the submission. Once all of the documents are received and confirmed, the review

timeline starts. Some health authorities have a structured approach and a system in place

tracking all of the applications while others do not have a structured approach.

The second step in the review process is to assign the different sections of the dossier to

different sub-committees. The application is usually broken up into two major sections,

Clinical, and Chemistry Manufacturing, and Controls (CMC). These two major sections

can be divided into several different sub-sections. Depending on the country and the

available capability and capacity, it could be one person reviewing each sub-section or

the entire section. If it is one person reviewing the entire section, then there will be a

great deal of dependency on the approvals of the more advanced countries. During this

stage of the review, it is normal for each reviewer to compile a list of questions and send

them to the applicant for additional clarification. In most cases, the health authority

representative would send the questions to the company representative in their country.

The company representative would then translate and send the questions to the global

regulatory person in the company who would then call on the technical experts to respond

to the questions. There are multiple communication channels and languages that the

information must travel through which adds complexity to the process. Once all of the

questions are answered and the health authority is satisfied, the reviewer(s) would then

provide a recommendation of continue / reject to the Registration Committee within the

health authority who then will issue the decision.

If the decision from Step 2 is to proceed, the health authority may ask the companies for

samples of the investigational product and reference (testing) materials. This step could

18

occur in parallel to Step 2. However, many countries prefer to wait to make sure they are

satisfied with Step 2 before investing resources on testing of a product that may not be

acceptable for other reasons. Since the application contains methods for testing the

product, the health authority will then either test the product samples or use an

independent analytical laboratory to test the product. If the results are within the

specifications set in the application, then a supportive statement is given to the

Registration Committee for approval. If the results are different or new peaks or

interference are seen in the chromatograms30

, questions are sent to the applicant for

clarification. The same steps would then follow as in Step 2 when sending

communication back to the technical experts in the company submitting the application.

The fourth and last step in the approval process is the pricing and labeling. The applicant

company can be requested to provide pricing data from countries where the product is

marketed to compare and generate a baseline. Other administrative information on

registration, marketing materials, and labeling in other countries may also be requested.

The Pricing Unit within the health authority proposes a price based on the information

provided by the applicant company and the country’s local guidelines. The Registration

Committee makes a decision based on the report from the sub-committees (e.g. scientific

assessment, lab analysis, and the cost analysis). While typically outside of the

registration process, the name of the product maybe different from country to country.

This is a negotiation the manufacturer will have with the health authority. The name

could be different from one country to another because the manufacturer may request it

30

Chromatograms are print-outs of chromatography results. Chromatography is a technique used to

separate and analyze mixtures of chemicals and results would show a peak for each chemical within the

mixture.

19

for promotional purposes or the health authority could request it because it may have a

certain connotation that may not be true once translated in the local language31

.

31

This information is based on the authors experience when working on products marketed in different

parts of the world. A tiered approach in this case would mean, as a hypothetical example, that a company

would set a price of $10 for the rich countries, $5 for medium income countries, and $2.50 for low income

countries with each tier having a different brand name. So the same product can be sold different countries

with different brand names.

20

Figure 5: Standard NDA Regulatory Review Process and Milestones32

32

Neil McAuslane and Tina Wang wrote the publication in March 2012 for CIRS titled: “New Drug

Approvals in ICH Countries: 2002 – 2011” http://www.cirsci.org/

Milestones

Official

submissi

on date

Process

Scientific

assessment

start time

Referred

to

Analytical

Laborator

y

Pricing

informatio

n

requested

Registrati

on

committee

decision Start of

queue

time

Submitted to

scientific sub-

committee

Validation

and Queue

Scientific

Assessment

Analysis of

samples

Collection of

pricing data

The authority

checks the

dossier to

ensure that

all

documents

are present

Company is

asked to

provide

additional

documentatio

n, if required

The validated

file waits in a

queue to be

picked up for

scientific

assessment

The dossier is assigned

to a member(s)of

the scientific sub-

committee for pre-

review and

completion of the

scientific report

form

Where necessary, the

company is asked

to provide further

information or

clarification

The report is referred

to the scientific

sub-committee

which makes a

recommendation

on whether the

product should be

registered

If the scientific

sub-committee

recommendation

is positive, the

product is sent to

the Lab for

analysis

Company is

notified and

provides samples

and reference

material for

analysis

Sample is

analyzed and Lab

issues a report on

whether it is

accepted

On acceptance of

the analysis the

company is

notified and asked

to provide pricing

information

Company

provides pricing

data from

countries where

the product is marketed

Other

administrative

information on registration in

other countries

may also be required

The pricing sub-committee

proposes a price based on the

information

provided and

guidelines

The Registration Committee makes

a decision based on the report from

the scientific sub-

committee, Lab and the pricing

sub-committee

Validation

& Queue

Scientific

assessment

Analysis of

samples

Collection of

pricing data

21

Chapter 2: Pharmaceutical Regulation

Medicines are probably as old as mankind and the concept about how their quality has to

be ensured has evolved gradually over time. For example, King of Pontus, Mithridates VI

(~120 BC), formulated a compound preparation which he called “Mithridatium” included

41 different ingredients and was used as a cure for many illnesses until as late as the

1780s33

. It wasn’t until 1540, when the Apothecaries Wares, Drugs and Stuff Act was

implemented in England, which required supervision of the manufacture of all medicines,

including Mithridatium34

. The Act was one of the first British laws to regulate medicines

and it instituted the appointment of four inspectors of “Apothecary Wares, Drugs and

Stuffs.” This could be viewed as the start of pharmaceutical regulations in England.

History of Pharmacopoeias, the official books of drug quality standards, probably dates

back to one of the proclamations of the Salerno Medical Edict issued by Fredrick II of

Sicily (1240), and ordered apothecaries to formulate medicines always in the same way –

forma curiae35

. The first Pharmacopoeias as we know them today were first seen in

Europe from the 16th century (e.g. the first Spanish Pharmacopoeia was issued in 1581).

The standards for the method of manufacture of Mithridatum were established in England

in The London Pharmacopoeia only in 161836

.

The modern medicines regulation started only after breakthrough progress in the 19th

century life sciences, especially in chemistry, physiology and pharmacology, which laid a

solid foundation for the modern drug research and development and started to flourish

33

Griffin JP, Shah RR. History of drug regulation in the UK. In: O’Grady J, Griffin JP, editors. The

Regulation of Medical Products. London: Blackwell BMJ Books; 2003 34

Ibid 35

Ibid 36

Rago, L., Santoso, B. Drug Regulation: History, Present and Future In: van Boxtel, C.J., B. Santoso and

I.R. Edwards, editors. Drug Benefits and Risks: International Textbook of Clinical Pharmacology, IOS

Press and Uppsala Monitoring Center, 2008

22

after World War II. Unfortunate events have catalyzed the development of medicines

regulation more than the evolution of a knowledge base. In 1937 over 100 people in the

United States died of diethylene glycol poisoning following the use of a sulfanilamide

elixir, which used the chemical as a solvent without any safety testing. This enabled the

establishment of The Federal Food, Drug and Cosmetic Act with the premarket

notification requirement for new drugs in 1938. However, in countries with poor

regulatory environment, even recently, medicines contaminated with diethylene glycol

have killed patients37

.

Over one hundred years ago, medicine in the United States was prescribed and dispensed

to patients by a pharmacist. The pharmacist was the one that determined which mix of

herbs or chemicals would treat an illness. In the United States, the first Pharmacopeia

was established in 1820 and the Philadelphia College of Pharmacy was established in

1821 producing trained pharmacists. However, in most of the world, there was little

governance back then of who would certify a person to be a pharmacist or approve the

'medicine' that was prescribed. It was a skill that was passed on from generation to

generation until the medical doctors developed an infrastructure to be in a position to

have the authority to prescribe the 'right' medicine to the patients. In the early 1940s,

chemical and pharmaceutical companies emerged as a strong industry that was able to

influence policy in the US and Europe38

.

37

In 1995-1996, 86 children in Haiti died after using Acetaminophen syrup that used Glycerin

contaminated with Diethylene Glycol. www.cdc.gov/mmwr/preview/mmwrhtml/00043194.html

In 2008 – 2009, 54 children under the age of 3 died from exposure to Diethylene Glycol that was used in

Acetaminophen based teething syrup in Nigeria.

www.cdc.gov/mmwr/preview/mmwrhtml/mm5848a2.html 38

Braithwaite, J., Drahos, P., Global Business Regulation, Cambridge 2000

23

In the nineteenth and early twentieth century in the West, pharmacy was a family

business, progressively subject to regulation by Pharmaceutical Societies. Pharmacists

would mix their own medicines. Patent-medicine peddlers travelled the countryside with

their wares or used local grocers as retail outlets. Some of these family businesses saw

the opportunities in internationalizing. They created new products by learning from

indigenous medicine in exotic places. Many large companies found the tropics

particularly rich sources of new biological materials in much the same way as they now

find them sources of genetic materials.

Regulation favored these international companies, crushing their small business

competitors39

. Pharmacopeias started more as recipe-books than as instruments of

regulation as long ago as the fifth century BC in Greece40

. Pharmacists, like cooks, could

experiment with the basic recipes and market their innovations. It was the medical

profession which slowed down pharmacist control over advice to consumers on drugs41

.

In most Western countries, the medical profession had successfully lobbied for laws to

require potent drugs to be sold only on the prescription of a licensed medical practitioner.

At the end of the twentieth century, developing countries are on the same trajectory;

though for most of the world’s poor population the local pharmacist remains a more

important source of therapeutic power and advice than the doctor or the global

pharmaceutical company.

After the thalidomide disaster of 1961, the medical profession lobbied states to require

more stringent safety testing and then to provide evidence of efficacy before a product

39

Ibid 40

McCoy, Alfred W. ‘Heroin as a Global Commodity: A History of Southeast Asia’s Opium Trade’, in

A.McCoy & A. Block (eds) War on Drugs: Studies in the Failure of US Narcotic Policy. Boulder,

Colorado: Westview Press, 1992. 41

Braithwaite, J., Drahos, P., Global Business Regulation, Cambridge 2000

24

was allowed to be marketed. In the 1970s, manufactures were also required in Western

nations, then progressively in developing counties, to comply with Good Manufacturing

Practices regulations, written by the US Food and Drug Administration (FDA) then

propagated by the World Health Organization (WHO).

By 1990 most pharmacists or ‘bathtub’ manufacturers could not afford the average of

$231 million for the research health regulators in advanced countries required before

allowing doctors to prescribe a new drug (D’Arcy & Harron 1999). Thus, regulation has

favored the strong – the doctors and the big corporations – over the pharmacists and

patent- medicine peddlers. In the poorest countries, where regulation is harder to enforce

and where consumers do not have enough money for the pharmaceutical industry or the

medical profession to be overly worried about chasing business, the global shift of power

to them from the local pharmacists and sellers of traditional medicines is yet to occur.

Current State of the Pharmaceutical Regulatory Framework:

The current system for making sure the medicine gets to the shelf on a pharmacy near

you is as follows: a pharmaceutical company must prove that the new medicine is safe

and efficacious and the company and produce millions of tablets that are within a set

specification – essentially make them over and over and over without any changes

(repeatability). The companies prove this by doing multiple clinical trials in local

markets and globally over the course of the drug development life cycle. All of the work

that needs to be done from discovering the drug to submitting an application for

marketing authorization is in the range of 12 – 15 years.

Here's where the fun begins! When a company is ready to submit their NDA to the

regulatory body, an application fee must be paid (for the United States, it is a little under

25

$2 million dollars). Once the NDA is submitted and the fee is paid, the FDA has 12

months to review and approve the application and issues a PDUFA date42

. There are

different timelines for special cases where there is an unmet medical need and the FDA

will grant a fast-track approval. For the purposes of this paper, we will only look at the

'regular' process for a new medicine across all of the countries. At the same time as the

NDA is submitted to the FDA, the pharmaceutical company can submit NDAs to other

countries and pay the associated fees. There is a small problem, the process isn't that

simple. There are only certain countries that can accept an NDA before any approvals

from major countries (i.e. US, EU, JP). The US, EU, and Japan are known to be leaders

in the pharmaceutical market. Once a major approval is granted by one of the major

markets, the pharmaceutical company will need to acquire a Certificate of Pharmaceutical

Product (CPP) from that government (this is the approval). The CPP can come from the

US, EU, Japan, or any developed country with a strong regulatory system. 75% of the

countries surveyed require a CPP at the time an NDA is submitted (some of the countries

will accept an application ahead of the CPP being available, but will not approve the

NDA until a CPP is submitted).

This means that 75% of the countries will have to wait at least 12-months, in the case of

the US NDA review time, before their government receives the NDA from a

pharmaceutical company. After the country receives the NDA, there are additional

requirements that each country must fulfill prior to approval. For example, China has

specific requirements that must be fulfilled prior to accepting an application (i.e. a

company must complete a local clinical study of 100 patients within China and have

42

PDUFA (Prescription Drug User Fee Act) was adopted in 1992 requiring drug and biologics

manufacturers to pay fees for product applications. The act also requires the FDA to use to issue a decision

date to the manufacturer.

26

favorable results). In addition to the requirements, many countries do not have the

capacity or capability to review the NDA and therefore delay the approval of the new

medicine, which in turn delays access of a new medicine to their market. Some countries

may never receive the application for not having a regulatory infrastructure or attractive

commercial market and therefore their patients may never have a choice to that medicine.

A significant amount of data has been gathered to show the difference in review time for

an NDA for each country as well as the amount of fees required per country (see chapter

4).

Because some pharmaceutical drug applications may not reach certain countries, patients

that require that particular novel drug may not have access to it because it is not approved

in their country. The alternative approach to receiving the drug would be through a

"compassionate use" or “single patient use” programs43

that are sponsored by the

manufacturing company or the patient would have to go to another country where the

drug is approved in order to obtain that drug (if they can afford it). The drug is not

approved in that country mainly because it is a low GDP country and therefore could not

afford the product or the sponsor company did not find the local market attractive. This

process is not available for every medicine – only to life-threatening diseases where no

alternative medicines exists or does not show efficacy in the named patient. There have

been examples of the sponsor company providing medicine to patients under the

‘compassionate use banner for several compounds. As an example, Schering Plough

43

There is no requirement for any manufacturer to conduct a compassionate use or single patient use

programs in all regions of the world. The studies are typically conducted in the regions where there is an

urgent need for the medicine and the study stops once the medicine is commercially available in that

particular country or region.

27

instituted a compassionate use program for its life saving drugs, Noxafil and Temodar,

for several years after gaining approval in the US and EU.

The current application process is specific for each country or region. In North America,

an application must be submitted to the US FDA. Another application is submitted to the

Canadian Health Authority (HA), and a third is filed with the Mexican HA. For Europe,

there are two different ways of submitting a new drug application: One is to submit a

single application to the EU HA (European Medicines Agency). The second is to submit

an application to each country individually and receive approval from each one of those

countries. For the most part, all of the applications have more or less the same

information. Multiple applications are prepared (extra work and cost for the

pharmaceutical companies), all the applications are reviewed by the various HAs

(overlapping use of government resources – time wasted), and most importantly delaying

access to patients around the world. The first EU option is better and is a potential model

that can be utilized globally. The focus on harmonization within the global technical

community has been on the CMC section as well as the clinical requirements section. In

the next chapter, we will discuss regional harmonization efforts and global technical

harmonization.

The process for Eastern Europe, Middle East & Africa (EEMEA), Asia Pacific (AP), and

South America has not yet been discussed but yield the same issues with duplicate

reviews. All of the countries and regions have some varying requirements (which

essentially are extensions of the first requirements already approved in the US, EU & JP)

which further delays access of the pharmaceutical product to the patients.

28

For example, countries like Egypt, Turkey, and the Philippines require that some type of

local manufacturing occur in their country. The regulations on this issue have been

evolving to include any part of the supply-chain (meaning manufacturing or packaging)

to the entire manufacturing and packaging process occurring locally.

Another example is Southeast Asian countries such as China, Japan, Taiwan, and South

Korea which require local clinical trial testing. This regulation requires companies who

wish to market their product in those countries to conduct clinical trials on a specific

number of local patients instead of counting on the global trials. In the past, a global trial

would be conducted and there may be patients from China, Japan, Taiwan, and South

Korea enrolled in the global clinical study. If enrollment is low at the clinical sites in the

aforementioned countries, and clinical sites in countries in Europe and the Americas are

enrolling at a high rate. This is a safety concern because those countries claim that their

patient population is different than that of Europe or the Americas (i.e. different digestive

system) and therefore the trial is not representative of their people. For example, if a

patient in Europe or the Americas is able to digest a certain amount of active

pharmaceutical ingredient (API) over a specified period of time, the concern is that a

patient in China or Japan may not be able to digest that same quantity. This leads to

overexposure of an active drug to the patient, which in some cases can cause renal failure

or impact other organs. If a manufacturer seeks to market the product locally, they are

required to conduct a separate local study that meets the countries’ requirements and

include the satisfactory results in the application. As a result, added cost, time, and

resources are needed to support a market launch in those countries. The outcome of the

local study could be that the dosage for countries in Southeast Asia differs (higher or

29

lower dose/strength) from other countries. If that were the case, this would mean that

additional drug development is required and the product for those countries would be

treated as a different product than that marketed in the rest of the countries.

Middle Eastern countries such as Jordan require additional post-market surveillance data

from manufacturers. For example, if a drug is approved in the US or Europe, the Jordan

health authority requires that the company submitting an application provide additional

data that shows that the commercially marketed product is safe after it has been marketed

to the general public for at least 12 months (CIRS 2011)44

. This requirement is in

addition to the clinical trials conducted by the manufacturer to prove the drug is safe and

efficacious to the first health authority that approved it.

The Institute of Medicine of the National Academies held a workshop in February 2013

to discuss regulatory harmonization and invited key stakeholders from governmental

agencies, non-governmental organizations, and Industry. The workshop held discussions

mainly on patient access to safe medicine and regulatory processes. The linkage between

safe medicine and regulatory systems is clear. Any product safety failure in any one

country can have ramifications around the world, and therefore the regulatory system is a

key factor in public health safety. The regulatory authorities in low and middle-income

countries often cannot perform all of the necessary responsibilities “…the FDA cannot do

its job well without substantive improvements in the capacity of its counterpart agencies

in the emerging economies” (IOM 2013). Specifically, the committee of regulatory

experts called for the sharing of inspection reports as an important first step in mutual

recognition and international regulatory harmonization.

44

CIRS 2011 Jordan Comparative Report.

30

Peter Honig, Global Vice President of Regulatory Affairs, AstraZeneca Pharmaceuticals,

summarized that harmonized standards in the pharmaceutical industry would45

:

Reduce costly duplication of effort;

Encourage sharing of experience and knowledge among regulators and scientists;

Require fewer clinical trials; and

Optimize use of limited resources

From the industry perspective, Honig highlights that harmonization would increase the

likelihood that a particular molecule will become a successful drug. Reduced

development time, less cumbersome approval processes across countries, and increased

speed to market are all important to companies. In addition, Honig believes that

harmonization would give patients faster access to new medicines and might lower the

costs of drug development, which could lower the price, making new drugs more

affordable in many more markets. An ongoing challenge for the industry and regulators is

to develop shared expectations regarding the use of adaptive trials, conduct of clinical

trials, acceptability of endpoints, and data transparency46

.

45

IOM 2013 46

Ibid

31

Persistent Barriers

Before clinicians can use a pharmaceutical product in a particular country, it needs to be

registered, approved, and marketed there. Honig cites numerous barriers to registration

that currently exist include47

:

China, Korea, and Taiwan require that a new drug be tested in subsets of their

population or in separate studies before it can be approved.

India, Mexico, and Vietnam require that specific numbers of their nationals

participate in clinical trials of the proposed drug.

Egypt, Turkey, and the Philippines require local manufacturing

Such requirements can create logistical difficulties in multiregional trials, when

researchers are required to allocate a certain number or percentage of trial slots to specific

groups of patients. In addition to the logistical issues, forcing a randomization code48

to

allocate certain percentages of patient groups to different centers may skew the study

results or hold up the completion of the study if patients do not enroll. Retaining

Investigational Review Board (IRB) approvals in each of the regions of the countries is

also a barrier as IRBs meet at a set schedule and have specific requirements about

submission of the protocols to be reviewed.

Challenges for existing harmonization initiatives

A large gap in regulatory capacity and expertise between emerging and developed

countries remains (IOM 2013). Programs developed by WHO helped governments in

less developed nations to be sure the drugs and medical devices imported into their

47

Ibid 48

Clinical trials use a randomization scheme where patients or drugs are randomized (or both). Different

treatment groups would receive active or placebo depending on the study design and randomization code.

Forcing a code means that the design of the code makes sure xx number of patients or drugs are used at a

defined time point.

32

countries are safe and effective, without requiring them to use limited resources to

replicate more developed nations’ regulatory infrastructures. This allows them to focus

on the issues of greatest local concern, such as the integrity of the supply chain.

Good Manufacturing Practice (GMP) inspections and re-inspections of manufacturing

and clinical trial sites can become burdensome. A global pharmaceutical company will

often have multiple inspectors coming in from different countries and from different

regulatory authorities in different regions. The incremental value of some of these

duplicate inspections becomes increasingly high and can be better utilized (IOM 2013).

Regulatory Perspective

Different countries take different approaches to medical products regulation, depending

on a number of factors. This is true even when they are geographically proximate,

operate under the same legal framework and rely on the same scientific processes and the

same data to make their decisions (IOM 2013). Some regulatory regimes may be more

risk-averse, while others may prioritize potential benefits. Whether they emphasize risks

or benefits may vary from one instance to another. As a result of these discordant

outcomes from regulatory decision making, patients in one country may have access to

medications that others do not have, which regulators may be hard pressed by patients,

providers, politicians, and the media to explain.

At a conference on regulatory harmonization in 2013, the Chair of the Dutch Medicines

Evaluation Board discussed the example of Avastin in a regulatory body taking this risk

benefit approach. As an example, the FDA revoked approval of Avastin for metastatic

breast cancer. Although the FDA originally approved the drug for this indication,

evidence that it did not extend life or improve the quality of life, while increasing the risk

33

of serious side effects, prompted the FDA’s decision. Yet, Avastin remains approved for

metastatic breast cancer in other countries! Such contradictory situations, some of them

widely publicized, can erode public trust in the system. However, the FDA’s public

report on the reasoning behind its decision is a model of balance and perspective. How

quickly do we forget? Fifty years ago, the FDA held up approval of thalidomide which

was proven to prevent thousands of birth defects that were found in EU after approval.

Schellekns and colleagues49

stated that regulatory systems should be assessed “in terms

of their ability to ensure patient safety, enhance public health, and stimulate innovation.”

Their effectives at achieving this latter aim are much in doubt, as the introduction of new

and innovative drugs has decreased sharply, despite rapid advances in biomedical

research. Schellekns and colleagues50

further stated, “Although the reasons for this

innovation deficit are not fully understood, many observers see the increasing demands of

the regulatory systems as one of the main drivers.” The regulators must take a step back

and ensure the right questions are being answered in assessing a new chemical entity.

Regulators use dossiers prepared by manufacturers in determining where to approve a

new drug. Problems associated with these dossiers are not infrequent. Typical problems

that can contribute to different regulatory decisions include the following51

:

Poor presentation: For example, the dossier presents data in a confusing way or

presents too much data, in which case the drug itself often receives a poor

assessment. Conversely, some dossiers may mask the data shortcomings by

strength of their presentations.

49

Schellekens, H., E. Moors, and H.G. Leufkens. 2011. Drug regulatory systems must foster innovation.

Science 332(6026):174-175 50

Ibid 51

Ibid

34

Coping with innovation: it may be difficult for regulators to assess a new concept,

so the default is to request more information, but whether such requests actually

produce an improved product is debatable. Some advanced therapies, including

gene therapies, may appear to regulators as too risky.

In the end, some of the variance in approval decisions across nations arises through the

dynamics of their individual review committees and their decision-making styles and

processes. Some nations base their processes on precise rules, whereas others base them

on principles. The latter approach gives greater flexibility to regulators but also reduces

the system’s predictability.

The labeling of a drug, which includes the indications for which its use is approved, can

vary among countries and change over time as new information is compiled. Sometimes

the number of indications is expanded and sometimes reduced; particularly if

complications arise that need to be more tightly controlled. A study of approaches used

by the FDA and EMA in the evaluation and approval of new cancer treatment indications

found real difference in the regulatory agencies’ wording nearly half (47%) of the

indications. However, the differences were clinically meaningful in only 10 of these

instances (Trotta et al., 2011).

Similarly, a study of differences in regulatory actions by the FDA and the European

Union related to biologicals appeared at first to suggest these differences were quite

large, but further analysis indicated that clinically relevant difference were much smaller

(Giezen et al., 2008). The more important feature was the timing in the two entities’

actions. The FDA was more likely to advise clinicians about potential problems sooner

than was the EU, and in some cases even to require a “black box warning” sooner. There

35

may always be differences in the ways people look at the data, how they weigh the

potential benefit or harm of specific products, and how they try to respond to their

populations’ unmet medical needs.

Evolution of harmonized regulations in the EU

The efforts to reduce differences in national drug regulation within Europe began in 1963,

when the European Commission (EC) hosted a conference of industry representatives,

doctors, and costumers to discuss harmonizing pharmaceutical laws52

. However, there

was disagreement over whether a drug should have proven “therapeutic potency” before

it could be approved. While doctors, pharmacists, consumers, and trade union

representatives insisted that this requirement was necessary, industry representatives at

the time refused to accept it53

. The result was stalemate.

In 1965, the EU issued its first pharmaceutical directive. It established baseline “criteria

for safety, quality and efficacy as preconditions for marketing authorization for new

medicine”54

. The directive also identified what constitutes medicinal products and

required member states to ensure that submissions of medicinal products to national

authorities were prepared and signed by experts with “the necessary technical or

professional qualifications”55

. These qualifications, however, remained undefined. In

addition, the efficacy test itself was limited: “Therapeutic efficacy would only be

considered lacking in a medicine which failed to produce pharmacodynamics results”56

.

52

Oraz, Luis, Kenneth Kaitin and Louis Lasagna. 1992. Pharmaceutical Regulation in the European

Community: Barriers to Single Market Integration. Journal of Health Politics, Policy and Law 17:859–861. 53

Vogel, David. The Globalization of Pharmaceutical Regulation. Governance: An International Journal of

Policy and Administration, January 1998 (11(1):1-22) 54

See Oraz et al. 55

Ibid 56

Hancher, Leigh. 1990. Regulating for Competition: Government, Law, and the pharmaceutical Industry

in the United Kingdom and France. Oxford: Clarendon Press.

36

Yet even this minimal requirement was implemented by only seven of the twelve

member states57

.

In 1974 the European Commission drafted a directive that permitted only “qualified

persons” to supervise drug production. But the Commission ultimately adopted such a

wide definition of qualified person (a medical professional, a veterinarian, a chemist, a

pharmaceutical technologist, or a biologist) that the directive became meaningless58

. Thus,

through the mid-1970s, virtually no progress had been made in creating a single market

for pharmaceutical products.

In 1975, the EC established the Committee for Proprietary Medicinal Products (CPMP)

with members requested from the regulatory authorities of the member states59

. The

CPMP was given the authority to review all drug applications by EU member states.

Applications would be examined for conformity to European Union safety, quality, and

efficacy standards, and the Committee would then issue an opinion on marketing

approval60

. However the CPMP’s role was only advisory. Member states maintained the

right to deny approval of an application. The multi-state procedure was intended not

only to generate confidence among member states about each other’s scientific

competence, but also to speed the approval of safe and effective drugs. Results were

mixed for this goal as well. Since the multi-state procedure had strict time limits (a state

had four months to evaluate a dossier that another state had already approved) member

states did give priority to multistate submissions. However, because member states still

57

Oraz et al. 1992, 854. 58

Ibid 59

Ibid 60

Buono, Teresa. Biotechnology-Derived Pharmaceuticals: Harmonizing Regional Regulation. Suffolk

Transnational Law Review 18:133, 1995.

37

carefully reviewed each submission rather than leveraging the initial state’s opinion, the

multi-state time limits were frequently violated (van de Gevel 1992). Only in France were

the time limits ever close to the intended plan. In Germany and the United Kingdom it

took an average of two years to review submissions, while in Italy, Spain, the

Netherlands, or Belgium it took three years or more (Howells 1992).

Although all member states agreed to accept test evidence gathered elsewhere in the

Community, there was no real progress towards mutual recognition of drug approval. As

the Committee’s chair noted in 1988, “Each concerned state seemed to conduct its own

assessment and raised its own objections . . . In practice there have been objections with

regard to each and every case dealt with under the Multi-State procedure.” He concluded

that “on the whole, the member states do not yet accept each other’s assessments”61

. The

implicated financial costs were significant. The Cecchini Report on “The Benefits of the

Single Market” estimated that the lost revenues of companies forced to wait over the

agreed 120 day limit was in a range between 100–175 million Euros, while multiple

registration requirements forced the applicants to employ extra staff at a cost of between

40 and 55 million Euros62

.

In 1987, unsatisfied by the outcomes of the multi-state procedure and committed to the

Community’s newly established goal of creating a single European market by 1992, the

EC took a radical new path. It approved a directive establishing a CPMP-administered

“Centralized Procedure.” This procedure was designed especially for biotech and high

technology products, since Brussels reasoned that it would be easier to harmonize

standards that had not yet been created than to require states to change the current

61

See Oraz et al. 62

Cecchini, Paolo. The European Challenge, 1992. Aldershot, England: Wildwood House, 1988.

38

procedures. The CPMP pooled scientific expertise in this new area of medical research in

order to create a Europe-wide consensus as to what represented good manufacturing

practices, adequate laboratory procedures and adequate evaluation criteria (Sauer 1991).

Once a manufacturer submitted its application, the CPMP would have seven months to

complete its initial review and evaluation; applicants were required to respond to both

CPMP and member states’ questions and comments within three months. The member

states then had up to one month to make their final recommendations, after which the

CPMP would send its report to all additional parties63

. While the CPMP’s report was

nonbinding, this new process was intended as the first move towards genuine regional

review and evaluation: “While fundamentally national, it [was] seen by the Commission

as a significant step in the direction of a single evaluation procedure applicable

throughout the Community”64

.

Following approval of the Maastricht Treaty on European Union (1992), which gave the

EU binding authority on some health care issues, the European Commission undertook

another major new initiative to harmonize national drug approval policies. It installed a

new regulatory institution, the European Medicines Evaluation Agency (EMEA) and two

new regulatory procedures65

. The EMEA consists of the CPMP, the corresponding body

for veterinary medicine, “a secretariat, an executive director, and a management board

composed of representatives of the member states, the Commission, and the European

Parliament.” The EU’s goal was to convert the relationship between national regulatory

63

Kanusky, Rosemary. 1994. Pharmaceutical Harmonization in the United States, the European Economic

Community and Japan. Houston Journal of International Law 16:665-707. 64

See Oraz et al. 65

Kingham, Richard, Peter Bogaert, and Pamela Eddy. 1994. The New European Medicines Agency. The

Food and Drug Law Journal 49:301–321.

39

authorities and those of the Union, to finally create a common regional review process for

pharmaceutical products.

An essential objective of the creation of a single European drug approval procedure was

to grow European-wide drug research and development, thus helping the industry to

“confidently continue to hold its place on the world stage in the foreseeable future66

. A

1995 communication from European Commissioner Martin Bangermann noted that over

the last two decades the share of all new medicines developed in the EU had declined

from half to one-third, while in the critical biotechnology sector, 65% of all patents were

American while only 15% were European67

. Increasing the efficiency of European drug

approval was regarded as a way of strengthening the effectiveness of this critical

industry.

The centralized procedure positioned the final regulatory approval at the Union level for

the first time. It permits manufacturers to submit applications directly to the European

Agency, which then refers them to the CPMP for review and evaluation. The latter is

required to issue its opinion within 210 days. If approval is denied, the drug’s sponsor

may file an appeal, which in turn must be reviewed within sixty days. Final approval rests

with the European Commission, which has ninety days to draft its own opinion. If the

Commission grants marketing authorization, it automatically becomes valid throughout

the EU for renewable periods of five years. The EMEA centralized approval process was

intended to be relatively fast, with application to final approval to take a maximum of ten

months68

. This is more than twice as fast as that of many member state drug regulatory

66

Pharmaceuticals: Carving Up Europe’s Drugs Industry. 1995. Economist August 26:57 67

Ibid 68

See Kingham et al.

40

agencies. To help the EMEA meet its own goals, applications can be accepted on CD-

ROM, and the agency’s questions and comments are to be provided by email69

.

As indicated earlier, the centralized procedure specifically targeted biotechnology drugs,

which must be approved by EMEA, as there is no national alternative. Biotechnology

was targeted because of its potential for economic growth and, because since it is such a

new field, individual states had not yet created their own testing infrastructures70

. As

one European official explained, “Global participation means we don’t have to keep on

reinventing the wheel. A new technique such as stereoisomerism, for instance, offers the

chance to put together a unified international approach before separate guidelines are

issued . . . The most obvious candidate here is biotechnology”71

. The centralized

application procedure was intended in part to benefit small firms with limited resources,

which were more likely to be found in the biotechnology sector. However, manufacturers

of any other pharmaceutical products may also choose to use the centralized procedure72

.

The Union also established a decentralized procedure that would be available to all

pharmaceutical products, except those produced through biotechnology73

. This

procedure, based on the principle of mutual recognition, was approved in 1993 and went

into effect on January 1, 1995. If a product has been approved for use in any member

state, its manufacturer could submit an identical scientific and technical application to

any or all other member states. It also must alert both the CPMP and the member state to

which its application was first submitted; the latter then is required to provide its

69

See Green 1995a 70

See Buono 71

Global harmonization on Pharmaceutical Regulations a Step Nearer. 1991. Pharmaceutical Business

News; Financial Times November 15. 72

See Kanusky 73

See Kingham et al.

41

assessment report to each country where recognition is sought. Each member state then

has ninety days to decide whether to recognize the first nation’s approval. It must do so

unless “there are grounds for supposing that the authorization of the medicinal product

concerned may present a risk to public health”74

. In cases of disagreement, member

states are urged to request the opinion of the CPMP.

While firms may still apply to individual member states for marketing approval, as of

January 1, 1998, any Member State which receives an application for a product which has

been approved by another member of the Union, must either accept that approval, or refer

the application to the CPMP for binding arbitration. Thus, pharmaceutical products will,

for the first time, be subject to mutual recognition, under the sponsorship of the EMEA.

Unlike the FDA, the EMEA’s role is a coordinating one; the actual processing of

applications is delegated to national regulatory agencies. This in turn means that the

agency must rely on member state authorities not only to administer applications in a

timely fashion, but also to apply similar criteria. The harmonization of national

requirements is even more important for the viability of the EU’s decentralized

procedure, since under this procedure a single national authority will be able to approve a

product for the entire EU. The inspection of manufacturing facilities and the certification

of the reviewers of applications could also vary between national authorities. However,

according to Fernand Sauer, the agency’s executive director, “after fifteen years of

harmonization we now have everything in place so that EMEA and all of the national

authorities practice exactly the same requirements” (Koberstein 1996).

Both new procedures promise to provide important benefits to manufacturers.

Streamlined approval allows cash flow to start sooner. “Successful new drugs earn $1

74

Ibid

42

million per day in global sales revenues. The European Union accounts for about 40% of

global sales” (Green 1995b). Also, faster drug approval will increase the actual life of

drug patents, increasing the potential value of future research75

. Since companies can

submit one application rather than fifteen76

, large firms may save up to $5 million

annually in national clinical staff and testing equipment77

. While the application fee for

submitting a drug to the EMEA is high a typical filing costs approximately 200,000

Euros—this is about half of what it would cost to pay all fifteen national fees. Centralized

approval also allows firms to use the same package inserts and make similar promotional

claims throughout the EU with the required languages.

Evolution of regulations in the US

The US NDA has a total of 19 sections that cover the lifecycle of a molecule from

discovery to commercialization. The bulk of the applications comprised of 2 major

sections – the Clinical section and the Chemistry, Manufacturing and Controls (CMC)

section. Within these 2 sections are clinical trial reports, drug stability reports,

toxicology, and drug-drug interaction reports, among many others. See appendix III for a

listing of all the sections within the NDA.

Reaching an NDA with 19 sections that generates tens of thousands of pages of data and

analysis did not occur over night. The regulations in the US started in 1820 when eleven

physicians met in Washington, D.C., to establish the U.S. Pharmacopeia, the first

compendium of standard drugs for the United States78

. Since then, many appointments

were made by Presidents within the administrations that followed through the 19th

75

Green, Daniel. 1995a. EU Body to Speed Up Approval of New Drugs. Financial Times January 26, 1995. 76

At the time Green wrote the article, there were only 15 member states in the EU. In 2014, there are 28

members states and all have adopted this process. 77

Ibid 78

See www.fda.gov

43

century until the actual implementation of the Food and Drug Act in 1906 that gave

legitimacy to the Food and Drug Administration. Regulations continued to evolve over

time throughout the 20th

century before reaching the 19 sections that make up the NDA.

The 20th

century brought many new requirements that manufacturers had to follow before

introducing a new drug to the market in the US and around the world (Table 1

summarizes some of the regulations introduced). Many of the changes generated a great

deal of controversy from manufacturers and from economists.

Table 1: Summary of laws and amendments introduced in the US regulations in the 20th

century

Year Law Purpose

1906 Food and Drugs Act Prohibited misbranded and adulterated food and

drugs in interstate commerce

1937 The Elixir Sulfanilamide

Incident

Use of diethylene glycol to dissolve Sulfanilamide

was toxic and resulted in over 100 deaths in more

than 15 states (234 of 240 gallons were recalled).

1938 Federal Food, Drug, and

Cosmetic Act

Tightened controls over drugs and food, included

new consumer protection against unlawful cosmetics

and medical devices, and enhanced the

government’s ability to enforce the law

1962 Kefauver-Harris

amendments

Thalidomide incidents – shift from economic to

safety concerns (proof of efficacy & FDA control

over clinical testing)

1983 Tamper-resistant

packaging regulations

Prevent poisonings such as deaths from cyanide

placed in Tylenol capsules

1992 Prescription Drug User

Fee Act

Requires drug and biologics manufacturers to pay

fees for product applications and supplements, and

other services. The act also requires FDA to use

these funds to hire more reviewers to assess

applications.

1997 Food and Drug

Administration

Modernization Act

Reauthorizes the Prescription Drug User Fee Act of

1992 and mandates the most wide-ranging reforms

in agency practices since 1938

2004 FDA Publishes

“Innovation or

Stagnation?”

Examines the path needed to bring therapeutic

products to fruition, and how FDA can collaborate in

the process

44

The Economics of Regulation

The theory of economic regulation was introduced by George Stigler in 1971 and further

built upon by Richard Posner and Sam Peltzman. The main argument is that regulation is

acquired by the industry and is designed and operated primarily for its benefit. Stigler

uses a simple model of regulation: A regulator (Congress or Parliament) faces special

interest pressure from producers and electoral pressure from consumers. The special

interest pressure is always more persuasive than others so producers always win.

Regulations are passed only for the benefit of large firms, not for the benefit or protection

of consumers.

This doesn't mean that regulators will be blatant about this. There are two ways to help a

producer: Via a direct subsidy or via protectionism. Subsidies aren't good--they

encourage new entrants into the market, so producers gain only a short-term benefit.

Protectionism, on the other hand, limits entry into the market--regulators favor this

method. So we see protective regulations like tariffs and occupational licensing fees

installed to help the large producers. Stigler's reasoning draws heavily on the point that

producers simply organize better.

Stigler79

proceeds from two primary premises:

1. The fundamental asset controlled by the state is the power to coerce. Any group

that can control how this power is used can profit.

2. Since we are self-interested actors, we will seek to get the state's coercive power

to support our interests. Efforts to do so, however, are costly.

79

Stigler, George. 1971. “The Theory of Economic Regulation” The Bell Journal of Economics and

Management Science. 2(1): 3-21.

45

Invariably, large firms win. Again, because they organize much better and are better

structured80

:

Large firms have high benefits from mobilizing. Since they are a small group, and

since they are fairly homogeneous, they have no difficulty with collective action

problems.

Small firms don't organize for political reasons because of collective action

problems: low potential benefits.

Consumers don't organize because the costs of doing so are high compared to the

benefits. Basically, consumers remain rationally ignorant.

This theory maybe applicable in the United States for most industries, but this may not be

the case for all of the regulations. Within the US, producers are able to influence policy

on pricing of pharmaceuticals, but not necessarily the regulatory review process and

timing. Outside the US, there are industrial groups in Europe, Japan, Canada and a few

other developed countries that serve a role in influencing policy. In the developing world,

there is an absence of up-to-date regulatory processes as well as industry groups. The

absence of both leaves room for corrupt actions or limitation on the ability to influence

the regulatory process.

In the next section, a review of the impact of the 1962 amendments to the Food, Drug,

and Cosmetic Act will show that the additional regulation limited the number of new

medicines introduced to the market thus limiting revenues for the producers and not

allowing patients access to potential new medicines. In this case, both parties lose and

the economic theory of regulation does not apply. Several economists studied the impact

80

Stigler, George. 1980. “An introduction to privacy in Economics and Politics” The Journal of Legal

Studies. 623-648.

46

of the new amendment on innovation, patient safety, and economic impact. Sam

Peltzman is one of the economists that sought to understand the impact of the added

regulation, specifically from the 1962 amendments requiring manufacturers to complete

efficacy trials. His initial study was to see what the impact of the regulations was on

innovation. The next section will give a summary of his findings.

The Peltzman review of the 1962 amendments to the Food, Drug, and Cosmetic Act:

Sam Peltzman has a serious issue with the regulations introduced in 1962 that required

pharmaceutical companies to conduct efficacy trials shifting the focus from economics to

safety. So much so, he wrote a couple of articles and a book with mathematical models

with results showing that the introduction of additional regulation has significantly

slowed down innovation in the pharmaceutical industry. Peltzman contends that the 1962

amendments decreased the amount of ‘safe’ drugs from reaching the market by more than

50% (Peltzman 1974). Although Peltzman did not set out to criticize the 1962

amendments prior to completing his study, and had given the benefit of the doubt to the

new regulations with favorable assumptions, he summarizes his final outcome to be that

the regulation cost the patients money and lives.

The 1962 amendments were introduced because of the Thalidomide incidents where a

number of people (17) died in the United States and much larger number of people in the

thousands died in Germany, the UK and other European countries. Thalidomide was an

approved drug in Europe, but not the United States. Patients from the United States were

able to get the drug as free samples from the doctor’s office (at that time, samples were

allowed to be provided to patients in anticipation of the drug approval). How can any

patient disagree with requiring a manufacturer to prove that the medicine said

47

manufacturer will market is indeed efficacious and safe? Peltzman suggests that

regulations were in place already to make sure manufacturers produced safe drugs

(proving efficacy came after the 1962 amendments). With that logic, one can concluded

that a manufacturer is able to produce a sugar pill and claim that it treats a certain kind of

illness without having to prove its efficacy. Playing the scenario out to an extreme,

patients would purchase the sugar pill, spend money on it, more importantly spend time

thinking this would help, only to realize that it was indeed not efficacious and did not

treat the intended illness. While it was safe to take (did not cause additional harm), the

patients may have missed the window to get treated with the right medicine that was

efficacious for that particular illness. Peltzman does credit the 1962 amendments with

attaining the goal of reducing the introduction (consumer waste) of ineffective drugs to

the market. However, based on his analysis in 1974, the costs in the process to have

clearly outweighed the benefits (Peltzman 1974).

In another 1974 publication, Peltzman cites an example of a drug that was approved in

the UK but was delayed approval in the US by 5 years (nitrazepam). The suggestion is

that 3700 deaths were seen in the US could have been averted had the drug been available

(without conducting the efficacy trials) because patients had to use other, less safe

sedatives and hypnotics81

. Peltzman later says “…greater risk taking is likely to yield net

benefits” (Petlzman 1974). Meaning that if we have several deaths for not conducting the

additional clinical efficacy trials and had more lives saved, that alone should be sufficient

to dismiss the additional requirements or lay blame on the requirements for the drop in

innovation!

81

Peltzman used pharmacologist William Wardell's estimate that because the relatively safe hypnotic drug

nitrazepam was not cleared for use in the United States until 1971, five years after it was available in

Britain, more than 3,700 Americans may have died from less safe sedatives and hypnotics.

48

From true numbers and an economics perspective, Peltzman may be correct. However,

the reality is that as technology evolves and scientists are able to learn more about new

chemicals because of improved equipment or testing methodology – the numbers don’t

tell the whole story. The science simply can’t be translated into numbers. As Vogel re-

iterates, regulatory harmonization is a key driver in promoting a higher number of new

medicines. The more regulatory authorities that have different requirements, the bigger

the hurdle to promote a new drug that must adhere to the different rules. As a result,

fewer innovative medicines are brought forward to the market. In the 1970s and 1980s,

new medicines introduced by European companies declined from half to a one-third and

only 15% of the biotechnology drugs (Vogel 2000).

49

Chapter 3: Regional Attempts to Close the Regulatory Gaps

Regional drug regulatory harmonization can be described by a number of initiatives

driven mainly by common socioeconomic needs and supported by global organizations

such as WHO and ICH. These initiatives are at different stages of development and

maturity, those including the more established countries being at a more advanced stage

than the less-resourced ones.

Marketing of pharmaceutical products is highly regulated because it involves several

ethical and human health and safety implications. As highlighted in earlier chapters, one

of the main obstacles to international approval of pharmaceutical products is that

different models for regulation of medicines exist in countries across the world. The

diversity of the regulatory requirements in different countries makes pharmaceutical

drugs’ applications and marketing very complex and costly. The added cost and

complexity often delays access of the public to critical and potentially life-saving drugs.

In addition, lack of respect for the Trade-Related Intellectual Property Rights (TRIPs)

agreement, strong price controls, and generic competition in the emerging and developing

countries remain a challenge for companies and could hinder marketing of innovative

drugs in these markets (Lakkis 2010)82

.

Historically, drug regulations in different countries evolved independent of each other,

affected mainly by local politics and economics, as well as the availability of resources

and the public health needs of the specific country. The substantial diversity in the

regulations, laws, and procedures of registering new pharmaceutical products, especially

between developed and developing countries, resulted in delayed patient access to

82

Maha Lakkis is an experienced regulatory affairs professional that has worked in industry for over 30

years and has published several articles on regulation in the pharmaceutical industry.

50

innovative medicines in the developing countries. Even with the acceptance of approval

by major regulatory authorities as evidenced by issuing a CPP (certificate of

pharmaceutical product), new drugs are still delayed by 1 to 2 or more years between the

time products are approved in developed and emerging countries (Lakkis 2010).

Recently, there has been an increased trend toward harmonization through increased

inter-country cooperation initiatives at regional and international levels, making borders

more open for trading among groups of countries with different regulatory, technological,

or financial backgrounds. In 2012, the FDA published a Global Engagement Report

where the FDA Commissioner, Margaret Hamburg, states “…Today we recognize that to

successfully protect U.S. public health, we must think, act, and engage globally. Our

interests must be broader than simply those within our own borders”83

.

In addition, the recent global expansion of the pharmaceutical industry by establishing

multinational manufacturing sites and moving towards marketing of new products in

international markets has made the need to standardize quality, efficacy, and safety

regulations stronger than ever. Therefore, global pharmaceutical regulatory

harmonization has become critically important for companies, international consumers,

and agencies; as a result, the different national regulatory agencies and industry

organizations started to cooperate more closely to harmonize and bring their regulations

closer to international standards (Lakkis 2010). The harmonization activities have been

often supported by intergovernmental initiatives at regional and interregional levels, as

well as by international agencies such as the World Health Organization (WHO) and the

International Conference on Harmonization (ICH) of Technical Requirements for the

83

The 2012 Global Engagement report is the first ever published report that stresses the need for global

cooperation on all fronts – regulation, imports/export, food supply chain, drug supply chain, and adding

FDA international posts to act like “embassies”.

51

Registration of Pharmaceuticals for Human Use (Vogel 2007). Harmonization of

pharmaceutical regulatory standards is important to reduce unnecessary duplication of

requirements, which delays access to new medicines. Practically, the regulatory standards

of safety and efficacy related to the review and availability of new medicines should be

essentially similar unless there are apparent regional and ethnic differences (Lakkis

2010).

International harmonization is characterized by a number of initiatives undertaken in

different regions of the world, driven mainly by common local or regional economic and

social needs. This chapter discusses some of the regional and global drug regulatory

harmonization initiatives. These initiatives are at different stages of development and

maturity, those including the more established countries being at more advanced stages

than the less-resourced ones.

European Harmonization

Europe was the first to lead the most advanced initiative for regional harmonization of

drug regulation. In the 1980s the European Commission (EC), now known as the

European Union (EU), led the first attempt to harmonize pharmaceutical regulatory

requirements, which was very successful in developing and implementing a structure for

harmonizing the drug regulatory laws and regulations across the region, leading to the

creation of a single market and promoting free circulation of pharmaceuticals within the

European member states. This was achieved through the establishment of the European

Council Regulation in July 1993, which resulted in the creation of the European Agency

for the Evaluation of Medical Products (EMEA) in 1995 (EMEA changed its name and

logo in 2010 to European Medicines Agency, EMA) to coordinate and facilitate the

52

European harmonization of pharmaceutical requirements84

. The creation of the EMEA

was a result of recognizing the increasing regulatory complexity, as well as the cost and

time required for development of new medicines; therefore, pharmaceutical companies

needed an effective and efficient regulatory environment within the EU to be more

effective in developing and marketing medicinal products. The most significant

achievement of the EMEA is the implementation of the Centralized Procedure in 1995,

which allows applicants to file one marketing authorization application that is assessed by

a centralized committee, the Committee for Medicinal Products for Human Use (CHMP),

which allows the approved products to be marketed in all EU member states85

.

At the same time, the option to use a specific national registration path in a particular

country only via national procedure, or within a certain number of countries via mutual

recognition procedure (MRP) or decentralized procedure (DCP), remains valid. The MRP

allows any national marketing authorization granted by any national authority of an EU

member state to support an application for mutual recognition by other member states.

On the other hand, in the DCP, the applicant selects a Reference Member State (RMS) to

lead the review procedure while being informed about other included states (Concerned

Member States) selected for marketing the product. Identical dossiers are sent to all

Concerned Member States as well as the RMS. After completing the review, the RMS

communicates its assessment results to the Concerned Member States, who can grant

marketing authorizations separately by the individual health authorities. However, any

Concerned Member State can disagree with the RMS assessment, which allows the

84

Council Regulation (EEC) No. 2309/93, July 22, 1993, laying down community procedure for the

authorization and supervision of medicinal products fro human and veterinary use and establishing a

European Agency for the Evaluation of Medicinal Products (OJ No L 214 of 24. 8.1993). 85

See Centralized Procedure: Directive (EC) No. 726/2004

53

applicant to pursue a dispute resolution phase. These procedures provide a more efficient

and cooperative approach than applying to several countries independently, especially

because these procedures include filing the same dossier, using identical specifications in

all involved countries8687

. These procedures include a scheme for involved states to

cooperate on post marketing surveillance of MRP and DCP products and to establish a

voluntary risk-based model of post market surveillance at the EU level, by sharing the

results of product testing with other participants, thus reducing duplication of procedures.

International Conference on Harmonization (ICH)

While the EC was working on harmonization in the EU, discussions started among

Europe, Japan, and the United States on possible harmonization among the three parties.

As a result, a unique tripartite harmonization project was initiated in 1989 through the

cooperative effort of the regulators and the industry associations in the three regions,

which led to the creation of the ICH in April 199088

. The ICH provided a multiregional

discussion forum for harmonization that led to significant progress in harmonizing

regulatory standards and technical guidelines for the registration of pharmaceutical

products (including new chemical entities and biotechnological products) and improved

the efficiency of global drug development (Lakkis 2010). The established ICH Steering

Committee (SC) consists of six voting members representing the regulatory bodies and

pharmaceutical industries of the three involved regions: it included members of the US

Food and Drug Administration (FDA) and the Pharmaceutical Research and

Manufacturers of America (PhRMA); Japanese Ministry of Health, Labor, and Welfare

(JMHLW), and Japan Pharmaceutical Manufacturers Association (JPMA), as well as the

86

See Mutual Recognition Procedure: Directive 2001/83/EC 87

See Decentralized Procedure: Directive 2004/27/EC 88

See International Conference on Harmonization www.ich.org

54

EU89

. The EU was initially represented by the EC and later transferred to the EMEA, in

addition to the European Federation of Pharmaceutical Industries Associations (EFPIA),

which represented pharmaceutical companies from 16 countries in Western Europe. The

ICH SC also includes nonvoting, non-ICH observer members representing WHO, the

European Free Trade Association (EFI'A) currently represented by SwissMedic (Swiss

Agency for Therapeutic Products), and Health Canada (Canadian Health Authority). The

Secretariat is provided by the International Federation of Pharmaceutical Manufacturers

Associations (IFPMA) (IOM 2013).

ICH’s mission is “to make recommendations towards achieving greater harmonization in

the interpretation and application of technical guidelines and requirements for

pharmaceutical product registration, thereby reducing or obviating duplication of testing

carried out during the research and development of new human medicines” (ICH, 2013).

The ICH was very successful in improving the development and licensing of new drugs

in the three regions in an efficient and cost-effective manner, by harmonizing regulatory

requirements and eliminating unnecessary duplication of clinical trials and animal testing

while maintaining the high standards of quality, safety, and effectiveness of the products.

The success of the ICH process has become a very good model of harmonization, with

over 50 guidelines for the technical requirements in four major categories of drug

development (quality, safety, efficacy, and multidisciplinary) and a dictionary of medical

terms. In addition, it has developed electronic standards, a common technical document

for electronic submission for registration of data on NDAs. The common technical

document includes guidance on formatting trial datasets and data elements, which

89

Ibid

55

facilitates review and enables industry to submit its data to different regulatory authorities

in a single format (Lakkis 2010).

Even though the ICH initially focused on developing guidelines and standards for the

ICH region, from the beginning, it maintained openness and transparency by keeping all

ICH guidelines and documents open to all authorities from all other countries at its

website. It also allowed representatives from non-ICH member states to attend ICH

plenary sessions. As a result, the effect of the successful ICH harmonization spread

beyond the three main ICH regions and its guidelines became increasingly perceived as

the gold standard for regulations in other countries, either by implementing them directly

or by following them when establishing their local regulations. Figure 6 shows an

illustration of how the ICH has evolved over the past 10 years from being a small player

connecting a small group of organizations to a major player driving harmonization across

many regions and implementing more than 50 guidelines.

Figure 6: Evolution of the ICH

56

Global Cooperation Group and Non- ICH Regional Harmonization Initiatives

The ICH successful model of harmonization triggered an international interest in the

process of harmonization, leading to several regional harmonization initiatives (RHIs)

that aimed to develop unified standards and guidelines on drug quality, safety, and

efficacy, across a defined regional group of non-ICH countries, with the aim to improve

access to essential medicines within their regions, using ICH procedures and guidelines.

Therefore, following the completion of the majority of its objectives in 1997, ICH SC

recognized the need to take a further step by expanding its efforts to support these

initiatives (Lakkis 2010). In 1999, it started to establish communication with interested

nonmember states and created a subcommittee designated as the Global Cooperation

Group (GCG) to serve as an information liaison between ICH and non-ICH countries and

to "make information on ICH activities and guidelines available to any country or

company that may be interested"90

. It also invited WHO to join, acting as a link between

ICH and non-ICH countries and regions. The GCG worked with WHO and other

international organizations to achieve acceptance and adoption of ICH guidelines in non-

ICH countries. Currently, WHO attends ICH SC meetings as an observer and has also

become an observer to the work of the GCG as a nonvoting member without decision

making capability. Currently, the most active regional harmonization initiatives include

the following:

- Asia-Pacific Economic Cooperation (APEC)

- Association of Southeast Asian Nations Pharmaceutical Product Working Group

- GCC’s Gulf Central Committee for Drug Registration

- PAHO’s PANDRH

90

See ICH Global Cooperation Group. http://www.ich.org/meetings/gcg-reports.html

57

- Southern African Development Community (SADC) and East African

Community (EAC)

- Individual countries, including Australia, Brazil, China, Chinese Taipei, India,

Republic of Korea, Russia, and Singapore

Asia Pacific Economic Cooperation (APEC)

APEC was established in 1989 among 21 countries of the Pacific Rim (Australia, Brunei

Darussalam, Canada, Chile, China, Hong Kong, Indonesia, Japan, Republic of Korea,

Malaysia, Mexico, New Zealand, Papua New Guinea, Peru, Philippines, Russia,

Singapore, Chinese Taipei, Thailand, United States, and Vietnam) with the objective of

enhancing economic growth and prosperity in the Pacific region91

. Some APEC member

states are also members of other RHIs such as ASEAN and the Pan American Health

Organization (PAHO). APEC, in partnership with ICH, set up a pharmaceutical

harmonization initiative in 2002 through the APEC Life Science Innovation Forum

(LSIF). LSIF established a Regulatory Harmonization Steering Committee and APEC

Network of Pharmaceutical Science to support cooperation on regulation of

pharmaceutical drugs and make medicines accessible to a larger population in the region.

The APEC network of pharmaceutical professionals and policymakers worked toward

harmonizing regulations among member states with the ultimate goal of developing a

single pharmaceutical market. APEC has collaborated with GCG to develop several

harmonization topics including92

:

1. Bridging studies

2. Stability in climatic zones of the APEC region

91

See Asia-Pacific Economic Cooperation www.apec.org

See: APEC harmonization center www.apec-ahc.org

See: APEC Health working group http://www.apechwg.org/portal/PortalHome.asp

See: APE Life Sciences Innovation Forum http://lsif.apec.org/ 92

Ibid

58

3. Clinical trials

4. GMP (good manufacturing practice) and GCP (good clinical practice)

implementations

5. Collaborative opportunity for common local clinical data that meet the regulatory

requirements for Korea, Japan, and China.

An APEC Harmonization Center (AHC) website is established under the authority of the

APEC-LSIF for supporting regulatory harmonization efforts and to provide a platform to

address and solve priority concerns of APEC member economies on regulatory

harmonization93

.

Association of South-East Asian Nations (ASEAN)

ASEAN was established among 10 participating Southeast Asian countries (Brunei,

Cambodia, Indonesia, Laos, Malaysia, Myanmar, Philippines, Singapore, Thailand, and

Vietnam) to promote regional economic unity and cooperation aiming to have a single

ASEAN market by 201594

. The initiative included a public health and pharmaceutical

harmonization scheme that includes developing harmonized guidelines for the regulation

of pharmaceuticals and a unified format of drug registration application, the ASEAN

CTD. It also includes a Mutual Recognition Agreement (MRA) on a post marketing alert

system to enhance pharmacovigilance capabilities by sharing information on marketed

medicinal products via one common reporting form and glossary, and supporting the

WHO Counterfeiting Taskforce Program as well as an MRA on bioavailability and

93

Ibid 94

See Association of Southeast Asian Nation http://www.asean.org/

59

bioequivalence by sharing relevant data and establishing one common list of comparators

for generics, similar to that of the WHO95

(Lakkis 2008).

The Gulf Cooperation Council (GCC)

The GCC, also known as the Cooperation Council for the Arab States of the Gulf, was

established in 1981 as a trade bloc among seven Arab states of the Persian Gulf (Bahrain,

Kuwait, Oman, Qatar, Saudi Arabia, United Arab Emirates, and Yemen as member in

Health Council) that share common economic and social interests. The GCC established

its patent office in 1992 and launched the GCC common market on January 1, 2008. In

1999, a pharmaceutical harmonization initiative was launched by a committee called Gulf

Central Committee for Drug Registration (GCC-DR), with the executive office being

located in Riyadh, Saudi Arabia. The GCCDR is composed of two members from each

participating country with the possibility of appointing two ad hoc nonvoting advisors

from its affiliates. The main objective of the GCC-DR is to coordinate health policies and

programs among the participating members via exchange of information, knowledge,

techniques, and expertise96

.

It is responsible for registration of pharmaceutical products, GMP inspection and

compliance, approval of quality control laboratories, and review of technical and post-

market surveillance reports. It also includes a Program of Bioequivalence Studies

according to a consolidated Registration Gulf Act. The GCC-DR adopted the ICH

guidelines as primary source for developing its own. It follows a centralized procedure

for drug registration with harmonized drug registration requirements and drug pricing for

95

See ASEAN Consultative Committee on Standards and Quality Pharmaceutical Product Working Group

http://www.asean.org/communities/asean-economic-community/item/accsq-pharmaceutical-product-

working-group 96

See Gulf Cooperation Council www.gcc-sg.org http://www.gcc-sg.org/eng/

60

all the member states. After centralized approval of a product, authentication and fee

payment should be followed in each of the member states as per their local policies97

.

The Southern African Development Community (SADC)

The SADC, headquartered in Botswana, is an intergovernmental organization created to

improve socioeconomic and political cooperation and integration among 15 southern

African member states (Angola, Botswana, the Democratic Republic of Congo, Lesotho,

Madagascar, Malawi, Mauritius, Mozambique, Namibia, Seychelles, South Africa,

Swaziland, United Republic of Tanzania, Zambia, and Zimbabwe). The ultimate

objective of SADC is to build a region with a high degree of harmonization and

rationalization to enable the pooling of resources and achieving collective self-reliance to

improve the living standards of the people in the region98

. The SADC Health Protocol for

Regional Cooperation and Integration was adopted in August 1999 to harmonize health

standards of quality, safety, and efficacy in the region, to ensure the efficient use of

resources and facilitate faster access to safe and effective medicines99

. Significant

progress has been made by SADC member states in the harmonization of guidelines and

there have been notable achievements on enhanced medicine access in a number of

member states. This was mainly achieved by strengthening regulatory capacities of

member states via "centers of excellence" that provide regional training on evaluation of

drug application dossiers, GMP compliance, and development of local GMP certification

to improve the quality of local productions, as well as training for qualification of testing

laboratories, development of a quality management program, and a market surveillance

97

See ICH GCC-DR working group http://www.ich.org/about/organisation-of-ich/coopgroup/gcc.html 98

See Southern African Development Community www.sadc.int and

http://www.sadc.int/themes/health/pharmaceuticals/ 99

See SADC protocol http://www.sadc.int/documents-publications/show/804

61

and monitoring program that helps fight counterfeit products. The major challenge to this

group is the disparity in the capabilities of the member states, as some members have

very limited resources.

The efforts of the SADC initiatives are part of a larger plan in Africa. There are several

other regional communities in Africa that are doing similar activities as that of SADC.

The East African Community (EAC) is a very active community that also has a working

group on drug regulatory harmonization for its five nation member states (Burundi,

Kenya, Rwanda, the United Republic of Tanzania, and the Republic of Uganda). There

have been setbacks in the initiatives due to limited funding, resources, technical

implementation of website, and lack of having a clear owner to the initiative. The SADC

and EAC working groups now fall under The African Medicines Regulatory

Harmonization (AMRH) Initiative was launched in 2010 to support African countries’

ability to review and approve medicines100

. This initiative provides holistic oversight for

the continent and better aligns resource needs across the continent as well as with

supporting organizations such as the WHO, World Bank, USAID, NGOs, and health

authorities such as FDA, EMA, etc. (ICDRA 2010).

Pan American Health Organization

PAHO is an international public health agency more than 100 years old that aims to

improve health and living standards of the countries of the Americas101

. It serves as the

WHO Regional Office for the Americas, being part of the United Nations system. The

PAHO/WHO Regional Office for the Americas established the Pan-American Network

for Drug Regulatory Harmonization (PANDRH) and its working groups to support drug

100

See East African Community http://www.eac.int/index.php 101

See Pan American Health Organization www.paho.org

62

regulatory harmonization processes102

. Participants in PANDRH include the national

authorities of countries in the Americas, various pharmaceutical interest groups, industry,

and academia. Current members include drug regulatory authorities of all PAHO member

states as well as representatives of the regional pharmaceutical industry associations such

as Latin American Association of Pharmaceutical Industries and Latin American

Federation of the Pharmaceutical Industry, members from academia, consumer groups,

professional associations, and representatives from the five sub-regional economic and

trade integration groups such as the Andean Community (Bolivia, Colombia, Ecuador,

Peru, and Venezuela), CARICOM (the Caribbean community), SICA (Central American

region with Costa Rica, El Salvador, Guatemala, Honduras, and Nicaragua),

MERCOSUR (Mercado Comun del Sur, including Argentina, Brazil, Uruguay, and

Paraguay), and NAFTA (North American Free Trade Agreement, including the United

States, Canada, and Mexico)103

. The harmonization activities of PANDRH include

establishing technical guidelines for harmonization of processes and standards to improve

drug quality, and programs for the strengthening of national regulatory agencies via

technical training programs. Twelve technical working groups (Good Manufacturing

Practices, Bioequivalence and Bioavailability, Good Clinical Practice, Drug

Classification, Counterfeit Drugs, Good Laboratory Practices. Pharmacopoeias, Medical

Plants, Drug Registration, Pharmacovigilance, Vaccines, and Promotion and Marketing)

were established and so far they have adopted eight final technical guidelines. In response

to these RHIs, the GCG endorsed new Terms of Reference in November 2003, to

establish collaborative partnership with RHIs and help them understand ICH guidelines.

102

See ICH PANDRH working group http://www.ich.org/about/organisation-of-ich/coopgroup/pandrh.html 103

Ibid

63

Consequently, GCG invited each of the RHls to designate permanent representatives to

attend the GCG meetings, and a GCG section was established on the ICH website as a

repository for documents and harmonization activities (Lakkis 2012, ICH).

The GCG membership currently includes one representative from each of the six ICH

parties on the Steering Committee, with observers (WHO, Health Canada, and IFPMA,

EFTA, and the ICH secretariat at IFPMA). It also includes a maximum of two permanent

representatives per regional harmonization initiative (APEC, ASEAN, GCC, PANDRH,

and SADC) to solicit and present the views of their regional initiative. In a 2005 meeting

in Brussels, GCG issued a mission statement: "to promote a mutual understanding of

regional harmonization initiatives in order to facilitate the harmonization process related

to ICH guidelines regionally and globally and to facilitate the capacity of drug regulatory

authorities and industry to utilize them." Since then, representatives from RHls have been

invited to listen to technical topics at the level of the Expert Working Groups,

Implementation Working Groups, Discussion Groups, and the SC (Lakkis 2010). In

October 2007, the ICH SC identified its goals:

1. To reduce country and regional differences in technical requirements that impact

the availability and cost of new medicines

2. To promote international movement of pharmaceuticals that are safe, effective,

and of high quality

3. To promote the conduct of clinical trials and data collection that meet

international standards

It also expanded the GCG membership and invited representatives from eight distinct

drug regulatory authorities (Australia, Brazil, China, Chinese Taipei, India, Russia,

64

Singapore, and South Korea) to attend the GCG meetings104

. These representatives are

granted the same access as RHIs at the biannual ICH meetings and invited to listen to

technical topics. In 2008, GCG's role expanded further, shifting from information sharing

to implementation issues and training.

Even though ICH has no legal mandate from the international community, recent

developments within GCG suggest that ICH activities are gaining wider international

interest and participating countries are following the ICH guidelines as the international

norm. It is noteworthy that GCG follows a set of principles when handling requests from

non-ICH parties: (a) ICH will not seek to impose its views on any country, region, or

company, but will serve as a source of information; and (b) ICH will provide non-ICH

member countries or companies with any document related to GCG initiative without

charge105

.

Role of the WHO in Global Regulatory Harmonization

WHO is an intergovernmental health international agency of the United Nations, with

193 member states. It is the only organization that has a legal international mandate from

member states to set global standards for the promotion and protection of public health.

WHO'S constitution specifically states that one of its functions is to "develop, establish,

and promote international standards with respect to food, biologics and pharmaceuticals

and similar products" to improve access to quality essential drugs, especially for poor and

vulnerable populations106

. WHO has been very active since the early 1970s in providing

guidance and assistance to regulatory agencies in the developing countries by

strengthening their national regulatory capacity and helping them to set up infrastructures.

104

See ICH Global Cooperation Groups http://www.ich.org/about/organisation-of-ich/coopgroup.html 105

Ibid 106

See World Health Organization http://www.who.int/about/en/

65

In addition, WHO played a critical role in driving harmonization of quality, safety,

efficacy, and nomenclature requirements on a global level. In addition, in collaboration

with a number of government agencies and individual experts, it developed several

practical guidelines on the quality and safety of drugs and established a mechanism to

review, modify, or adopt ICH guidelines as appropriate (Rago and Santoso 2008). WHO

provided a supportive mechanism for approval in countries with less regulatory capability

by implementing the WHO Certification Scheme and encouraging the use of CPPs issued

by developed regulatory authorities as a valid document to support approval in

developing or underdeveloped countries. WHO also supported global harmonization by

convening the International Conference of Drug Regulatory Authorities (ICDRA) every 2

years since 1980, which provides an important forum for communication, coordination,

and collaboration among drug regulatory authorities of WHO member states107

. The

objectives of the ICDRA are to (a) promote collaboration among drug regulatory

authorities, (b) reach a consensus on matters of common interest, (c) facilitate timely and

adequate exchange of information, and (d) discuss issues of international relevance. Over

the years, ICDRA became a very effective network and a forum where senior drug

regulators can discuss issues, exchange ideas, and set direction for future development108

.

It also played an instrumental role in guiding regulatory authorities from all WHO

regions by determining priorities for action in national and international regulatory issues.

During the 1989 conference, plans began to develop global pharmaceutical regulatory

standards by addressing critical regulatory issues, and at the thirteenth ICDRA

107

See WHO ICDRA http://www.who.int/medicines/areas/quality_safety/regulation_legislation/icdra/en/ 108

Ibid

66

preconference109

, it addressed an important issue, better medicines for children, and

recommended that WHO convene and collaborate with the global pediatric group of

regulators to identify priorities and treatment guidelines needed for neonates, and

establish mechanisms to support drug development for children. ICDRA also

recommended that WHO should do the following:

1. Promote adoption and implementation of the WHO Model Registration Package

as minimum information requirements for product registration

2. Produce guidance and draft regulation for managing confidentiality issues among

regulatory authorities

3. Undertake joint assessment of selected applications using the WHO Model

Registration Package

4. Foster the development of regional, collaborative post-market surveillance and

pharmacovigilance systems to monitor the quality, safety, and efficacy of health

products

5. Establish formal mechanisms for the exchange and use of regulatory information

among all authorities to strengthen capacity and maximize efficiencies

Summary

Drug regulations have evolved over time in response to increasing scientific knowledge

and the complexity of the pharmaceutical industry. The pharmaceutical industry is

becoming increasingly multinational, and pharmaceutical companies are now focusing on

global drug development to take advantage of the opportunities in the rapidly expanding

new markets in Asia, Latin America, the Middle East, and Africa. This globalization

109

See 13th

ICDRA conference

http://www.who.int/medicines/areas/quality_safety/regulation_legislation/icdra/presentations13thICDRA/e

n/

67

trend highlights the need for a new strategic approach of pharmaceutical harmonization.

Globalization of standards has become a necessity and a goal for many groups of

neighboring countries in several regions to reduce unnecessary and duplicative

requirements, rationalize time and costs, and create a transparent regulatory process,

resulting in improved access to medicines both domestically and globally. Global

harmonization became possible thanks to the success of ICH in providing very advanced

and comprehensive guidelines and global standards, and in supporting the development

of RHls leading to harmonized regulatory standards. Drug regulatory harmonization is

beneficial to the pharmaceutical industry as well as consumers: therefore, harmonization

efforts are supported by governments as well as the pharmaceutical industry: PhRMA

groups as well as EFPlA are playing a constructive role in supporting global

harmonization via international regulatory affairs groups that act as a liaison with local

and multinational regulatory and trade organizations to advocate pharmaceutical industry

views (IOM 2013). Substantial progress has been achieved over recent years throughout

the world, but much remains to be done. However, a complete international

harmonization of drug regulations and establishing one set of international standards is

still far from reality, and no single regional initiative can be considered an ideal model for

international application and implementation, but at least a new trend is moving in the

right direction toward global harmonization.

68

Chapter 4 – Methodology and Data Analysis

Introduction

The pharmaceutical industry is often viewed as a money-making machine with market

cap of $1.2 trillion in 2011! It’s also a very expensive business to be in costing major

pharmaceutical companies $4 – 7 Billion USD in R&D investment every year (IMS

2012)110

. These figures are specific to innovative companies manufacturing new

molecular entities, not generics. Still, if the industry’s revenues are so high, why aren’t

all the medicines accessible to everyone around the world? That’s a simple question with

a very complicated answer. This chapter will attempt to address this question in

reviewing economic, social, political, sociopolitical, and education data from over 100

countries around the world. The data will be analyzed to identify the relationship of the

NDA review period (time) with the cost of application (cost), life expectancy (LE), health

expenditure per person, and literacy rate. Said differently, time data is linked to assess

the impact on quality of life, which is linked to the ability of access to medicine. The

quality of life is determined by the life expectancy in a given country, GDP per capita,

literacy rate, and health expenditure per person.

The research strategy is to relate NDA review cycle time to economic, political, and

quality of life characteristics. The hypothesis being that a country with a stable

democracy will have a structured regulatory process which leads to timely access of

newly developed medicine. Additionally, in order to have a structured regulatory process

with defined timelines, a country must have a high literacy rate. The more educated the

people, the stronger the regulatory structure, the longer life expectancy – which is linked

to access to medicine based on a set review time and cost.

110

IMS Health publication in 2012 on the pharmaceutical industry

69

Data, Methodology, and Regression results

Detailed description of the variables

The research strategy for this paper is to relate review time of a new drug application

(time) to:

1. Quality of life indicators (life expectancy and physicians per 1000 people),

2. Economic status (GDP per capita, Health Expenditure per person, openness of

economy, and cost of the applications), and

3. Political status (Democracy score, State fragility index, size of government)

The NDA review time and fee were collected by conducting a survey with regulatory

professionals working for a global pharmaceutical company and therefore represent real-

time data. The data was supplemented and confirmed from a review conducted of the

IDRAC reports published by Thompson Reuters Regulatory Intelligence unit. The

IDRAC reports provided data on 69 countries based on their research. Ministries of

health generally do not publish timelines on their websites or in any other outlet. As a

result, the data collected from the survey will be the only published data available for all

the countries included in this study. The data submitted by the regulatory professionals

was provided in units of either days or months. All of the time data was converted to

days for consistency in the analysis. The cost of the application fee (cost) was provided

in either local currency or USD. All of the cost data was converted into USD by using

the Oanda currency converter website111

.

111

Oanda is a currency converter website that is broadly used - http://www.oanda.com/currency/converter/.

Monetary conversions made in March – May 2012.

70

GDP per capita and Life Expectancy, Physicians per 1000 people, and Health

Expenditure per person were collected from the World Bank world development

indicators report of 2012112

. The variables are defined as:

- GDP per capita: basic economic indicator and measures the level of total

economic output relative the population of a country.

- Life expectancy at a specific age: average number of additional years a person

of that age could expect to live if current mortality levels observed for ages above

that age were to continue for the rest of that person’s life. In particular, life

expectancy at birth is the average number of years a newborn would live if current

age-specific mortality rates were to continue.

- Physicians per 1000 people: number of physicians per 1000 people in a given

country. The number includes general practitioners as well as specialists.

- Total health expenditure: sum of public and private health expenditures as a

ratio of total population. It covers the provision of health services (preventive and

curative), family planning activities, nutrition activities, and emergency aid

designated for health but does not include provision of water and sanitation. Data

are in current U.S. dollars.

The two political stability indicators, Democracy Score and State Fragility Index, were

collected from the Polity project113

. The Polity conceptual scheme is unique in that it

examines concomitant qualities of democratic and autocratic authority in governing

institutions, rather than discreet and mutually exclusive forms of governance. This

perspective envisions a spectrum of governing authority that spans from fully

112

The World Bank publishes “World Development Indicators” for all the countries around the world 113

The polity project is supported by the Political Instability Task Force, Societal-Systems Research Inc.,

and Center for Systemic Peace http://www.systemicpeace.org/polity/polity4.htm

71

institutionalized autocracies through mixed, or incoherent, authority regimes (termed

"anocracies") to fully institutionalized democracies. The democracy score captures this

regime authority spectrum on a 21-point scale ranging from -10 (hereditary monarchy) to

+10 (consolidated democracy). The Polity scores can also be converted to regime

categories: a three-part categorization of "autocracies" (-10 to -6), "anocracies" (-5 to +5

and the three special values: -66, -77, and -88), and "democracies" (+6 to +10). The

Polity scheme consists of six component measures that record key qualities of executive

recruitment, constraints on executive authority, and political competition. It also records

changes in the institutionalized qualities of governing authority. The Polity data include

information only on the institutions of the central government and on political groups

acting, or reacting, within the scope of that authority.

The State Fragility Index and Matrix list all independent countries in the world in which

the total country population is greater than 500,000 in 2010 (164 countries). The Fragility

Matrix scores each country on both Effectiveness and Legitimacy in four performance

dimensions: Security, Political, Economic, and Social, at the end of the year 2010. Each

of the Matrix indicators is rated on a four-point fragility scale: 0 “no fragility,” 1 “low

fragility,” 2 “medium fragility,” and 3 “high fragility” with the exception of the

Economic Effectiveness indicator, which is rated on a five-point fragility scale (including

4 “extreme fragility”). The State Fragility Index, then, combines scores on the eight

indicators and ranges from 0 “no fragility” to 25 “extreme fragility.” A country’s fragility

is closely associated with its state capacity to manage conflict, make and implement

public policy, and deliver essential services while progressing development114

.

114

ibid

72

Open K and KG were extracted from the Penn World Table – Center for International

Comparisons. The Penn World Table provides purchasing power parity and national

income accounts converted to international prices for 189 countries/territories for some or

all of the years 1950-2010115

. Open K is the economy openness at 2005 constant prices

(%). It is a calculated number scoring the openness of economy with regard to trade.

Trade is the sum of exports and imports of goods and services measured as a share of

gross domestic product. KG is the Government Consumption Share of PPP Converted

GDP Per Capita at 2005 constant prices. It is a calculated number identifying the size of

the government consumption. General government final consumption expenditure

(formerly general government consumption) includes all government current

expenditures for purchases of goods and services (including compensation of employees).

It also includes most expenditures on national defense and security, but excludes

government military expenditures that are part of government capital formation.

The data sets described in the section above was collected and uploaded to a spreadsheet.

Linear regression analysis was conducted using Ordinary Least Squares method (OLS)116

.

Definitions and summary statistics for important variables used in this study are

presented in Tables 3 and 4. The summary statistics show extremes between nations

across all of the variables. Health expenditure per capita, for example, ranges from the

low number of just under $15 in the Democratic Republic of Congo and the highest

number being over $8200 in the United States. Physicians per 1000 people ranged from

115

Alan Heston, Robert Summers and Bettina Aten, Penn World Table Version 7.1, Center for

International Comparisons of Production, Income and Prices at the University of Pennsylvania, Nov 2012.

https://pwt.sas.upenn.edu/php_site/pwt_index.php 116

OLS is a method for estimating the unknown parameters in a linear regression model. This method

minimizes the sum of squared vertical distances between the observed responses in the dataset and the

responses predicted by the linear approximation.

73

the low of 0.008 doctors/1000 people in Tanzania to 6.167 doctors/1000 people in Greece.

GDP per capita ranged from $105 in the Democratic Republic of Congo to Luxembourg

at $52,000.

The database collected for this research is unique in that it compares variables that had

not been looked at in the past and therefore the analysis is unavailable. The list of review

time for NDA approval for all of the countries was not found in any one publication. All

of the regression in Table 4 shows the regression results of the log time variable on the

rest of the 9 variables. Regression analysis was not conducted on all variables at the

same time. The first regression results show that the Log of GDP per capita yields a

statistically significant result when regressed alone. The second regression shows that

Log of GDP still yields a statistically significant result when regressed with Cost. In fact,

both variables, Log GDP per capita and Cost, yield statistically significant results. When

regression analysis is conducted with more than one other variable, Log GDP does not

yield a statistically significant result. Health Expenditure, Physicians per 1000 people,

Fragility, and Life expectancy consistently yielded a statistically significant result

indicating that faster drug approval means higher health expenditures and longer life

expectancy values. The results also suggest that faster drug approvals are occurring

because of the higher literacy rate. The results for Literacy rate indicate that for every 4 –

5 days the review time is reduced, there is a 1% increase in literacy rate. Being

conservative and taking the 5 days per 1%, the results indicate that Literacy Rate in a

country would increase by 20% if the review time was 100 days less.

Life expectancy seems to be the most important variable because it implies that:

74

- Countries with lower review time and high NDA fee will have higher life

expectancy

- Population would live 10 more years more if the approval review time is

reduced by 128 days

One of the surprises of the results is the significance level of the NDA fee in regression

results with multiple variables. It was expected that the NDA Fee (cost) would be

significant when correlated with any other variable. The results in Table 4 indicate that

cost is significant when regression conducted with time and one other variable only – this

was expected. However, when 2 or more variables were introduced, cost was significant

at the 10% level – this is the unexpected result. As an example, if NDA fee is increased

by $100k, the NDA review time would only improve by 27 days. The expectation was

the cost would play a bigger role in the review time and other variables. The second

surprise was the insignificance of the democracy score. In all the regression analysis, the

democracy score was consistently insignificant. Fragility, on the other hand, was

consistently significant. This can be interpreted to mean that a country must have

stability, but not necessarily an open democracy to have access to medicine in a timely

manner. This was a very important result when looking at broken states versus an

autocratic regime. The broken state will surely not have access to medicine, but the

autocratic regime seems to have better access as long as there is stability in the country.

Summary of results

Log GDP per capita was statistically significant when regression analysis on log time was

conducted alone or with cost only. When regression analysis conducted with more than

one variable, it was no longer significant. This result was also unexpected as one of the

75

points in the hypothesis was that richer countries would have more stability and less

review time. The results indicate that GDP per capita plays a critical role when it is a

standalone variable, not when combined with other variables. Cost, as discussed earlier,

was also a surprise. While statistically significant in many of the regressions, the actual

value of the improvement on review time was minimal. For example, we would expect

to see $100K in increased cost that for 27 – 40 days improvement in approval time.

Health Expenditure yielded statistically significant results across all data sets. However,

the actual financial value of the Health Expenditure was not very high, similar to the

results of cost. Open K was not statistically significant in any of the regressions. This

result implies that having an open economy does not necessarily yield a faster drug

approval. Physicians per 1000 people had a clearly significant result in the regression

analysis with time alone. Statistically significant results were also seen when regression

analysis was conducted for Physicians per 1000 people with cost, Health Expenditure,

Open K, and LogGDPPC. However, regression results of Physicians per 1000 people

with Fragility or Life Expectancy does not yield a statistically significant result.

Regression analysis of Literacy rate with cost yielded statistically significant results.

However, regression analysis did not yield statistically significant results of Literacy rate

with life expectancy. An example of the result between Literacy and cost is that for

$100k increase in NDA fee, the review time is reduced by 93 days. This result is three

times better than the result seen with cost alone where $100k increase in NDA fee only

yielded 27 – 40 days in a shorter review cycle. Therefore, higher Literacy rate

contributes to reducing the review time when the same increase in cost is applied.

76

Fragility alone gives statistically significant results. Fragility yields statistically

significant results in regression analysis with all other variables except when Life

Expectancy is included in the analysis. Fragility yields statistically significant results in

regression analysis with Democracy score and Open K. This result implies and confirms

that a stable and open democracy that trades with other governments will have a lower

review cycle and therefore access to medicine. Life Expectancy yields a very high

statistically significant result in regression analysis alone with time or with GDP per

capita. Life Expectancy yields statistically significant results in regression analysis with

cost and physicians per 1000 people as well as Open K and Health Expenditure. Life

Expectancy does not yield statistically significant results in regression analysis with

Fragility and GDP per capita. These results indicate that Fragility and Life Expectancy

are equally important to access to medicine. This result makes sense as stability is

required in order to have access to medicine which would support an increase in Life

Expectancy. The results indicate that lack of education in a given country leads to a

limited number of educated people (low literacy). Infrastructure in a country cannot be

built without stability and an educated population. Therefore, capabilities cannot be

established to review the complex applications for new medicines and economic growth

is limited.

A summary of regression results using the Cox Hazard model can be found in Table 5.

The results of the Cox Hazard117

model yielded that Life Expectancy is a very important

117

Cox Hazard is a survival model that relates the time that passes before some event occurs to one or more

covariates that may be associated with that quantity of time. In a proportional hazards model, the unique

effect of a unit increase in a covariate is multiplicative with respect to the hazard rate. For example, taking

a drug may halve one's hazard rate for a stroke occurring, or, changing the material from which a

manufactured component is constructed may double its hazard rate for failure. Other types of survival

models such as accelerated failure time models do not exhibit proportional hazards. The accelerated failure

time model describes a situation where the biological or mechanical life history of an event is accelerated.

77

variable giving a result of nearly 1.04. This means that life increases by 10 years with 128

days less of approval time. This result is in line with the OLS outcomes discussed earlier.

Using the Cox Hazard survival model was an experiment to see what the results would

yield and were added to this section.

In addition, the same analysis for the results in Table 4 was conducted using time, instead

of log time, as the dependent variable. The data are summarized in Table 6 in Appendix

VI – regression analysis data with dependent variable time. Cost is not statistically

significant with time but is statistically significant with log time as shown in Table 4.

Adjusted R2 is not as good with time as the dependent variable as seen in table 6.

Adjusted R2 is higher in all the regression analysis results when log time is the dependent

variable (Table 4). This suggests that, because countries’ review time can be divided into

groups, applying the log function to time better represents the industry trends.

78

TABLE 2

VARIABLE DEFINITIONS

Health Expend*

Health expenditure per capita in a given country (USD)

Open K**

Percentage of economy openness at 2005 constant prices

Cost+

Fee charged by a health authority in a country to review a new drug

application (USD)

LG GDP PC** Log of GDP per capita in a given country

Dem Score***

Classification of a country based on regime; Full Democracy (10),

Democracy (6 to 9), Open Anocracy (1 to 5), Closed Anocracy (0

to -5), Autocracy (-6 to -10), and Failed (-66 to -77)

Phys / 1000* Number of physicians per 1000 people in a country

Frag. Index***

Fragility Index scores each country on both effectiveness and

legitimacy in four performance dimensions: Security, Political,

Economic, and Social, at the end of the year 2010. The higher the

number (0 – 25), the more fragile the country

Life Expectancy*

Average life expectancy in a given country (combined

male/female) (years)

KG** Government consumption share of PPP converted GDP per capita

at 2005 constant prices

Time+

Amount of time needed to review a new drug application (days)

118

Sources:

* World Bank

** Penn World Table

*** Polity Project – Center for System Peace

+ Survey results and research

118

Accelerated review of life saving medicines had a shorter approval time.

79

TABLE 3

MEANS AND STANDARD DEVIATIONS OF IMPORTANT VARIABLES FOR

COUNTRIES STUDIED FOR THE YEARS 2010 – 2011

VARIABLE

MEAN STANDARD

DEVIATION OBSERVATIONS (# OF COUNTRIES)

Health Expend

1205.14 1928.70 125

Open K

91.34 56.92 129

Cost (USD)

57335.3 198029.5 129

LG GDP PC

7.94 1.62 124

Dem Score

4.91 10.35 128

Phys / 1000

1.97 1.48 106

Frag. Index

7.71 6.03 124

Life Expectancy 70.21 9.60 128

KG 9.62 5.59 129

Time (days) 744 265.39 129

80

TABLE 4

RESULTS OF REGRESSION ANALYSIS OF LOG APPROVAL TIME ON COUNTRY ECONOMIC AND POLITICAL

CHARACTERISTICS ACROSS 129 COUNTRIES, 2010 - 2011

Variables OLS

(t-Stat) (1) (2) (3) (4) (5) (6) (7)

LG GDP PC -0.077 -0.056 -0.017 -0.018

(-3.73)* (-2.85)* (-0.88) (-0.93)

Cost -7.52E-07 -2.82E-07 -2.81E-07 -3.08E-07 -3.05E-07

(-4.75)* (-1.70) (-1.71) (-1.80) (-1.80)

Health Expend -0.00010 -0.00013 -0.00011 -9.98E-05 -0.00011

(-5.50)* (-8.65)* (-6.37)* (-5.11)* (-5.98)*

Open K -0.00033 -0.00031

(-0.66) (-0.63)

Constant 7.15 7.03 6.81 6.69 6.68 6.85 6.71

(42.38) (44.52) (45.08) (203.15) (204.40) (42.44) (125.01)

Adjusted R2

0.095 0.23 0.39 0.37 0.38 0.38 0.38

# of observations 124 124 121 125 125 121 125

*statistically significant

81

TABLE 4 (continued)

Variables OLS

(t-Stat) (8) (9) (10) (11) (12) (13) (14)

LG GDP PC 0.0020 0.013

(0.092) (0.63)

Cost -3.04E-07 -3.21E-07 -3.16E-07 -2.64E-07 -2.55E-07 -2.45E-07 -2.67E-07

(-1.79) (-1.94) (-1.88) (-1.62) (-1.59) (-1.51) (-1.69)

Health Expend -0.00010 -7.40E-05 -7.36E-05 -6.10E-05 -7.48E-05 -7.68E-05 -6.35E-05

(-5.77)* (-3.71)* (-3.50)* (-2.70)* (-3.55)* (-1.51) (-2.99)*

Open K -0.00032 2.03E-06 6.25E-05 0.00029 0.00017 0.00028 0.00030

(-0.63) (0.0040) (0.12) (0.55) (0.33) (0.53) (0.59)

Dem Score -0.0019

(-0.70)

Phys / 1000 -0.078 -0.080 -0.034

(-3.30)* (-3.22)* (-1.29)

Frag. Index 0.021 0.022 0.024 0.013

(2.86)* (3.75)* (3.62)* (1.77)

Life Expectancy -0.010

(-2.26)*

Constant 6.72 6.78 6.75 6.49 6.45 6.32 7.21

(122.94) (107.47) (39.34) (54.46) (71.72) (29.87) (20.83)

Adjusted R2

0.38 0.44 0.43 0.48 0.46 0.46 0.48

# of observations 125 106 103 103 121 117 121

82

TABLE 4 (continued)

Variables OLS

(t-Stat) (15) (16) (17) (18)

LG GDPPC 0.013

(0.58)

Cost -2.66E-07 -4.62E-07 -4.82E-07 -2.58E-07

(-1.64) (-3.14)* (-3.30)* (-1.57)

Health Expend -5.59E-05 -6.23E-05

(-2.43)* (-2.72)*

Open K 0.00034 0.00060 0.00042 0.00029

(0.64) (1.07) (0.77) (0.54)

KG 0.0015 0.00054 0.0027

(0.23) (0.084) (0.43)

Phys / 1000 -0.023 -0.051 -0.051 -0.032

(-0.82) (-1.90) (-1.91) (-1.20)

Frag. Index 0.017 0.031 0.029 0.020

(2.05)* (4.01)* (4.03)* (2.75)*

Life Expectancy -0.0064

(-1.18)

Constant 6.94 6.22 6.38 6.46

(17.24) (24.53) (48.26) (49.14)

Adjusted R2

0.48 0.45 0.44 0.48

# of observations 103 100 103 103

*statistically significant

83

TABLE 5 (Cox Hazard)

Variables (Hazard Ratio)

(z value)

(1) (2) (3)

Dummy<60 0.58 0.60

(-4.19)* (-3.91)*

Cost 1.000001 1.000001

(3.38)* (3.04)*

Life Expectancy 1.04

(5.56)*

# of observations 129 129 128

Dummy: countries with life expectancy under 60 is 24 of 129

*statistically significant

84

Discussion:

Life expectancy was the most important variable yielding statistically

significant results in both the OLS and Cox Hazard models. While the

focus of this study has been using OLS regression, Cox Hazard analysis

was another attempt at generating results to confirm outcomes. According

to the data and results, people in a given country would live 10 years

longer if the approval time is reduced by 128 days. While that sounds

simple, it is not. In order for a country to reduce the cycle time for

reviewing all of the drug applications, it will need medical experts

(literacy rate) who are capable of reviewing the new drug applications. In

order to have experts review the applications, a country must be stable

(fragility index) in order for the political system to develop and thrive

(democracy score). Once a stable political system is in place, economy

will grow because a strong political system attracts investments. If a

country sustains political stability and investments are made in the

country, the people living in the country can thrive in education (literacy

rates) and obtain high-income jobs (GDP per capita).

Based on the above, a conclusion was made that all of the indicators were

linked and cannot be separated. While the OLS results did show that one

variable alone could yield statistically significant results, the true result

was when there are several variables were analyzed at the same time. A

surprise variable in this analysis was cost. It was expected that cost would

play a big role, maybe even bigger than life expectancy or literacy rate. In

85

reviewing the data, cost ranged from zero to $2M. Qatar, for example, is

one of the wealthiest countries in the world (high GDP per capita) does

not have a fee for a new drug application. This input could potentially

skew the result. The United States, on the other hand, has a cost of $2M

for an application – nearly 4 times as much as the next highest application

fee. Removing Qatar alone or the United States alone out of the dataset

would probably not change anything. Removing both extremes (groups of

nations) could potentially change the output. However, removing the

United States risks other variables such as Literacy rate and life

expectancy to be impacted negatively which could change the previously

seen results.

The other story to be told here is that the US, EU, Japan, Australia,

Canada with firm regulatory infrastructure and high cost deliver approvals

on time, every time. This group of nations is in the upper tier for all of the

variables. They have a high GDP per capita, high literacy rate, excellent

democracy and fragility scores, and have a similar timeline for review and

approval of new drug applications.

86

Chapter 5

Conclusion

This work began with a basic question. Why does it take a long time

before a new drug is available to patients in all of the countries? The

initial thought was to look at the general regulations, determine the gaps,

leverage or develop a governance structure that would allow for a global

approval, and the question would be answered. We now know it is not

that simple. There was a complex history of the evolution of regulation

globally and how the regulation is impacted by the economics and

political landscape in a country or a region. While many questions were

addressed in this paper, there is still more work to be done.

Studying the different variables in each country and reviewing the results

of the regression analysis made things abundantly clear. The issue is not

that governments do not want to have medicines available to their

populations. The issue is not necessarily that people cannot afford it,

although that is concern. The issue is that in order for access to medicine

to reach the top of the agenda, more pressing issues need to be addressed.

For example, having a stable government with an educational system in

place is a must before reestablishing or enhancing a drug regulatory

process. Cost of the application fee was thought to be a clear reason why

some countries were unable to have access to the medicine. After the

regression analysis, it’s very clear that a stable government with an

educational system is a must and how much money is paid for the NDA is

87

not as critical, because without a stable government and an educated

people, the NDA fee becomes insignificant. By taking the unique

approach of evaluating multiple variables across the political and

economic systems more insight to the key factors is gleaned as discussed

in Chapter 4 which can be further studied and built upon.

The term Global Affairs suggests that there are multiple subjects being

discussed. The results of this paper imply and confirm that a

multidisciplinary approach must be taken in order to address current

global issues. Economics are dependent on the politics and politics are

dependent on the economic policies. History and international law are

closely connected with the financial and political systems. Development

indicators are an outcome and a direct result of the interdependencies of

these subject fields. The results of this study confirm this statement in that

the regulatory system cannot be studied independently of the political and

economic development indicators. If a single system is studied

independently, the results will not be sufficient. This is confirmed in the

results shown in Chapter 4. Cost is statistically significant on its own, but

not when analyzed with other factors.

Furthermore, economists and policy makers must become active

participants with the healthcare and regulatory professionals. In the

research conducted, it was evident that industry leaders and regulatory

bodies (ministries of health) are heavily involved in the current

discussions on regulatory issues, harmonization, and access to medicine.

88

What is recommended is having additional government representatives

from other departments sit at the table to make it a more fruitful

discussion. The current process has Global Cooperation Groups (GCG)

focused on pharmaceutical regulations work in conjunction with bigger

regional organizations. A GCG is under the umbrella of APEC, ASEAN,

SADC, EAC, PAHO, GCC and so forth. The GCG sits in a silo, discusses

the issues, and reports them to the bigger groups. This is a problem.

Instead of having regulators and industry professionals recommending

policy, economists and politicians need to inform these policies, not be

informed. A holistic approach is needed to support a viable approach that

can yield a realistic path forward.

At the National Academies Institute of Medicine (IOM) conferences as

well as WHO’s International Conference on Drug Regulatory Authorities

(ICDRA), for example, executives from all the major industry players as

well as high level officials from all of the regulatory bodies were present

discussing key issues in pharmaceutical regulation. However, there were

no representatives from the ministries of commerce, foreign affairs,

interior, or education. This is a key learning from this study – that is

having the right people at the table to inform policy making. Based on the

research conducted, there is a lack of ‘other’ voices on the table when it

comes to global regulatory harmonization and policy making.

Regulatory / policy implications

89

The hypothesis was that countries with a higher GDP per capita, higher

democracy score, and lower fragility index will have a higher fee and

shorter time for review. In other words, if the state is stable, infrastructure

will be in place to review the NDA in a timely manner for a set cost that

the applicant will be able to afford because the return on investment for

the application will yield substantial gains. The results of the analysis

from Chapter 4 confirm part of the hypothesis. The cost of the NDA is

statistically significant on its own. But it is not statistically significant

when analyzed with other variables. Fragility of a country and life

expectancy results were the strongest among all of the variables and

statistically significant.

Based on the regional harmonization efforts, a recommendation for a

potential regulatory model that will help alleviate the resource constraints

and enable faster approval of medicines globally is possible. The model

would involve an agreement where shared resources from neighboring

countries and regions work together to approve a new drug. As a result,

the approval would be instantly applied to all of the countries involved.

The current harmonization efforts in leveraging resources are only on a

regional level. In order for a model to be successful, a broader effort must

be made in utilizing expertise from more developed regulatory systems to

support the underdeveloped systems. The support cannot be only

consulting, but rather more hands on of coaching and training. As

indicated earlier, the cooperation and collaboration has to be across

90

multiple disciplines for this to be a success. This means that ministries of

education need to be involved in the educational system of a developing

country. This means that other ministries (commerce, interior, foreign)

must be involved to understand the impact of economic and development

indicators on a successful model. The model used to develop a

harmonized process in the European Union can be a significant starting

point. The EU had multiple players around the table discussing how the

harmonization model could play out. They were successful not only

because of the right players, but also because they had a head start in a

developed regulatory model and strong economies. The developing world

is lacking behind on education, economic and political status, and

therefore, the effort to bring about a globally harmonized process will be

an extremely challenging project.

Future Work:

This study focused on the regulation of new innovative pharmaceutical

products. There are different classifications for all of the medicines and

therefore this study did not cover all of them. There are regulations for

generic medicine, regulation for Over the Counter (OTC) products,

regulation for Biologics, and regulation for conducting clinical trials. The

work conducted for this paper could be expanded into these other fields

and confirm if the same results would be reached. Generics and OTC

products follow a different model in terms of review time and cost, and

therefore, may not have similar outcomes. The regulations of these

91

different medicines can be the same as pharmaceutical products studies

and it can be different. In some cases, such as the OTC products, there

may be limited or no regulation versus a regulatory process that is exactly

equivalent to the pharmaceutical products studied in this paper.

Deciphering which regulatory system applies in the different countries and

applying the statistical analysis will be challenging and results would be

very interesting to see. Generics also follow a different process that the

innovative pharmaceutical products studied in this paper. The correlation

between the variables for generics could be clearer and more

straightforward. However, because generics are also prescription

products, the CPP requirement would still apply and therefore a similar

trend in some of the variables should be expected. Once policy makers,

regulators, and industry leaders can clearly understand where the biggest

opportunity (based on data) lies, there will be more traction on a

multidisciplinary approach to global regulatory harmonization.

92

Bibliography

1. Abraham, J., Helen Lawton Smith. 2003. Regulation of the Pharmaceutical

Industry. New York : Palgrave Macmillan.

2. APEC Harmonization Center. www.apec-ahc.org

3. Asia-Pacific Economic Cooperation. www.apec.org

4. Berry, Ira, Martin, Robert P. 2008. The Pharmaceutical Regulatory Process.

New York: Informa Healthcare.

5. Braithwaite, John. 1994. Prospects for Win-Win International Rapprochement

of Regulation. In Regulatory Co-operation for an Interdependent World.

Paris: OECD.

6. Braithwaite, J., Drahos, P. 2000. Global Business Regulation. Cambridge.

7. Buono, Teresa. 1995. Biotechnology-Derived Pharmaceuticals: Harmonizing

Regional Regulation. Suffolk Transnational Law Review 18:133.

8. Cecchini, Paolo. 1988. The European Challenge, 1992. Aldershot, England:

Wildwood House.

9. Center for Innovation in Regulatory Science: http://www.cirsci.org/

10. Council Regulation (EEC) No. 2309/93. July 22. 1993. Laying down

community procedures for the authorization and supervision of medicinal

products for human and veterinary use and establishing a European Agency

for the Evaluation of Medicinal Products (OJ No L 214 of 24. 8. 1993).

11. Davies, Norman. 1997. Europe: A History. London: Pimlico.

12. D’Arcy, P.F. & D.W.G Harron (eds). 1991. Proceedings of the First

International Conference on Harmonization. Belfast: Queen's University of

Belfast.

13. Drug Regulation. Economist November 16:84. 1991.

14. European Medicines Agency www.emea.eu

a. Centralized Procedure: Directive (EC) No.726/2004.

b. Decentralized Procedure: Directive 2004/83/EC.27/EC

c. Mutual Recognition Procedure: Directive 2001/

15. Food and Drug Administration www.fda.gov

16. Global harmonization on Pharmaceutical Regulations a Step Nearer.

Pharmaceutical Business News; Financial Times November 15, 1991.

17. Giezen, T.J., A. K. Mantel-Teeuwisse, S.M. Straus, H. Schellekns, H. G.

Leufkens, and A.C. Egberts. 2008. Safety-related regulatory actions for

biologicals approved in the United States and the Euripoean Union. JAMA

300(16):1887-1896.

18. Grabowski, H., J. Vernon, and L. Thomas. 1988. Estimating the Effects of

Regulations on Innovation: An International Comparative Analysis of the

Pharmaceutical Industry. Journal of Law and Economics 21 (1): 133-163.

19. Grabowski, Henry and John Vernon. The Regulation of Pharmaceuticals;

Balancing the Benefits and Risks. American Enterprise Institute Studies in

Government Regulation

93

20. Green, Daniel. Fast-track to Approval. Financial Times April 24, 1997.

_____. 1995a. EU Body to Speed Up Approval of New Drugs. Financial Times

January 26.

_____. 1995b. European Medicines Evaluation Agency: Fast-track Approvals

Service. Financial Times April 25.

21. Griffin JP, Shah RR. 2003. History of drug regulation in the UK. In: O’Grady

J, Griffin JP, editors. The Regulation of Medical Products. London: Blackwell

BMJ Books.

22. Gulf Cooperation Council. www.gcc-sg.org

23. Hancher, Leigh. 1990. Regulating for Competition: Government, Law, and the

pharmaceutical Industry in the United Kingdom and France. Oxford:

Clarendon Press.

24. Howells, J. Pharmaceuticals and Europe 1992: The Dynamics of Industrial

Change. Environment and Planning 29:33–49, 1992.

25. Institute of Medicine (IOM). 2013. International Regulatory Harmonization

Amid Globalization of Drug Development: Workshop Summary. Washington,

DC: The National Academies Press.

26. International Conference on Harmonization. www.ich.org

27. ICH Global Cooperation Group. www.ich.org/cache/compo/276-254- 1 .html

28. ICH Global Cooperation Group. Terms of Reference, November 2003.

www.ich.org/LOB/media/MEDIA341Zpdf

29. The International Conference of Drug Regulatory Authorities, Bern,

Switzerland, September 16-19, 2008.

30. Jordan, David. 1992. International Regulatory Harmonization: A New Era in

Prescription Drug Approval. Vanderbilt Journal of Transnational Law

25:471–505.

31. Kahn, Alfred, 1988. The Economics of Regulation: Principles and Institutions.

Cambridge, Mass.: MIT Press.

32. Kanusky, Rosemary. 1994. Pharmaceutical Harmonization in the United

States, the European Economic Community and Japan. Houston Journal of

International Law 16:665-707.

33. Kingham, Richard, Peter Bogaert, and Pamela Eddy. 1994. The New

European Medicines Agency. The Food and Drug Law Journal 49:301–321.

34. Knight, Kenneth, George Kozmetsky and Helen R. Baca. 1976. Industry

Views of the Role of the Federal Government in Industrial Innovation,

University of Texas at Austin.

35. Koberstein, Wayne. 1996. Fernand Sauer: Tending the Garden.

Pharmaceutical Executive 16:36–7.

36. Lakkis, M. 2010. Global and Regional Drug Regulatory Harmonization

Initiatives. Drug Information Journal, 44:289-297.

37. Lakkis M. 2008. The ASEAN harmonization scheme: another CTD-another

challenge to the industry. Regul Focus. November: 39-43.

38. Lakkis M. 2008. The WHO certification scheme and international

pharmaceutical product marketing. Regu1 Focus. June: 34-37

39. Life Sciences Innovation Forum. www.apec.org/apec/documen ts-

reports/life-sciences-innovation-forum/

94

40. McAuslane, Neil and Wang, Tina. 2012. “New Drug Approvals in ICH

Countries: 2002 – 2011”. Center for Innovation in Regulatory Sciences

(CIRS).

41. McCoy, Alfred W. 1980. Drug Traffic: Narcotics and Organized Crime in

Australia. Sydney: Harper & Row.

42. McCoy, Alfred W. 1992. ‘Heroin as a Global Commodity: A History of

Southeast Asia’s Opium Trade’, in A.McCoy & A. Block (eds) War on Drugs:

Studies in the Failure of US Narcotic Policy. Boulder, Colorado: Westview

Press.

43. Oraz, Luis, Kenneth Kaitin and Louis Lasagna. 1992. Pharmaceutical

Regulation in the European Community: Barriers to Single Market Integration.

Journal of Health Politics, Policy and Law 17:859–861.

44. Pan American Health Organization. www.paho.org

45. Peltzman, S. 1974. Regulation of Pharmaceutical Innovation. Washington,

DC: American Enterprise Institute.

46. Peltzman, S.1973. An Evaluation of Consumer Protection Legislation: The

1962 Drug Amendments, 81 Journal of Political Economy 81 (5) 1049-1091.

47. Peltzman, Sam. 2010. “Regulation and the Natural Progress of Opulence”

Institute of Economic Affairs 30 (2) 33-39.

48. Peltzman, Sam. 1993. “George Stigler’s contribution to the Economic

Analysis of Regulation” Journal of Political Economy. 101 (5): 818-832.

49. Peltzman, Sam. 1974. Regulation of Pharmaceutical Innovation; The 1962

Amendments. American Enterprise Institute for Public Policy Research,

Washington, D.C.

50. Peltzman, Sam. 1976. “Toward a more general theory of regulation” The

Journal of Law and Economics. 211-240.

51. The Penn World Table : http://www.rug.nl/research/ggdc/data/penn-world-

table

52. Pharmaceuticals: Carving Up Europe’s Drugs Industry. 1995. Economist

August 26:57.

53. The Polity Index: http://www.systemicpeace.org/polity/polity4.htm

54. Rago, L., Santoso, B. 2008. Drug Regulation: History, Present and Future In:

van Boxtel, C.J., B. Santoso and I.R. Edwards, editors. Drug Benefits and

Risks: International Textbook of Clinical Pharmacology. IOS Press and

Uppsala Monitoring Center.

55. Sauer, Fernand. 1991. Harmonization of Biotechnological Regulations in the

European Community. In Drug Biotechnology Regulation: Scientific Basis

and Practices, eds., Yuan-yuan H. Chiu and John L. Gueriguian. New York:

M. Dekker.

56. Schellekens, H., E. Moors, and H.G. Leufkens. 2011. Drug regulatory systems

must foster innovation. Science 332(6026):174-175

57. Southern African Development Community. www.sadc.int

58. Southern African Development Community strategy

http://www.sadc.int/themes/health/pharmaceuticals/

59. Stigler, George. 1971. “The Theory of Economic Regulation” The Bell

Journal of Economics and Management Science. 2(1): 3-21.

95

60. Stigler, George. 1980. “An introduction to privacy in Economics and

Politics” The Journal of Legal Studies. 623-648.

61. Trotta, F., H.G. Leufkens, J. H. Schellens, R. Laing, and G. Tafuri. 2011.

Evaluation of oncology drugs at the European Medicines Agency and U.S.

Food and Drug Administration: When differences have an impact on clinical

practice. Journal of Clinical Oncology 29(16):2266-2272

62. van Boxtel, C.J., B. Santoso and I.R. Edwards. 2008. Drug Benefits and

Risks: International Textbook of Clinical Pharmacology, IOS Press and

Uppsala Monitoring Center.

63. Van de Gevel, A.J.W. 1992. The Pharmaceutical Industry. CEPSWorking

Paper No. 42

64. Viscusi, W. Kip, Joseph E. Harrington, Jr., John M. Vernon. 2005. Economics

of regulation and antitrust. Cambridge, Mass.: MIT Press.

65. Vogel, David. 1995. Trading Up: Consumer and Environmental Regulation in

a Global Economy. Cambridge: Harvard University Press.

66. Vogel, David. 1998. The Globalization of Pharmaceutical Regulation.

Governance: An International Journal of Policy and Administration. 11(1): 1-

22.

67. Vogel, Ronald. 2007. Pharmaceutical Economics and Public Policy, New

York: Pharmaceutical Products Press.

68. World Health Organization. www.who.int

69. World Health Organization. International Conference of Drug Regulatory

Authorities.

www.who.int/medicines/areas/quality_safety/regulation_legislation/icdra/en/i

ndex.html

70. WHO ICDRA Meetings

http://www.who.int/medicines/areas/quality_safety/regulation_legislation/icdr

a/en/

71. WHO Medicine Expenditure Report –WHO/EMP/MIE/2011.2.6

http://apps.who.int/medicinedocs/documents/s18767en/s18767en.pdf

96

Appendix I: List of countries included in the survey with data collected

97

Country / Region Cost

(USD)

CPP Time for approval

(days)

Algeria 3300 Y 995

Argentina 10000 Y 795

Armenia 5600 Y 730

Australia 200,000 N 300

Austria 50000 N 365

Azerbaijan 2500 Y 730

Bahrain 148 Y 815

Bangladesh 100 Y 485

Belarus 6000 Y 545

Belgium 11470 N 540

Benin 960 Y 1095

Bhutan 100 Y 485

Bolivia 5000 Y 665

Bosnia and Herzegovina 5500 Y 635

Botswana 104.8 Y 1095

Brazil 40000 Y 1095

Bulgaria 5500 N 365

Burkina Faso 1055 Y 1095

Cameroon 960 Y 1095

Canada 350000 N 365

Central African Republic 870 Y 1095

Chad 680 Y 1095

Chile 2500 Y 445

China 11000 Y 730

Colombia 4500 Y 395

Costa Rica 3600 Y 730

Croatia 5000 Y 595

Cyprus 2000 N 365

Democratic Republic of

Congo

950 Y 1095

Denmark 25000 N 365

Dominican Republic 3600 Y 730

Ecuador 2500 Y 545

Egypt 2500 Y 730

El Salvador 3600 Y 730

Ethiopia 800 Y 730

Finland 15000 N 365

France 70000 N 400

98

Country / Region Cost

(USD)

CPP Time for approval

(days)

Gabon 1055 Y 1095

Georgia 2500 Y 485

Germany 150000 N 365

Ghana 4500 Y 730

Greece 350000 N 365

Guatemala 3600 Y 730

Guinea 655 Y 1095

Honduras 3600 Y 730

Hong Kong 351 Y 815

Hungary 14,384 Y 619

India 1000 Y 905

Indonesia 3000 Y 575

Iran 5000 Y 1095

Iraq 165 Y 1095

Ireland 25000 N 730

Israel 5528 Y 730

Italy 120,000 N 600

Ivory Coast 680 Y 1095

Jamaica 75 Y 545

Japan 350000 N 365

Jordan 2500 Y 730

Kazakhstan 4000 Y 730

Kenya 1000 Y 730

Korea 3696 Y 1095

Kuwait 356 Y 1095

Kyrgyzstan 500 Y 730

Lebanon 2567 Y 1095

Libya 20000 Y 730

Luxembourg 200 N 540

Madagascar 750 Y 1095

Malawi 420 Y 730

Malaysia 1350 Y 730

Mali 1055 Y 1095

Malta 150 N 365

Mauritania 861 Y 1095

Mauritius 677 Y 1095

Mexico 6300 Y 730

Moldova 1100 Y 730

Mongolia 0 Y 730

99

Country / Region Cost

(USD)

CPP Time for approval

(days)

Montenegro 2300 Y 1095

Morocco 15000 Y 1085

Mozambique 610 Y 730

Namibia 555.1 Y 1095

Nepal 100 Y 485

Netherlands 350000 N 365

New Zealand 10000 N 460

Nicaragua 3600 Y 730

Niger 1055 Y 1095

Nigeria 1590 Y 730

Norway 350000 N 365

Oman 195 Y 730

Pakistan 1100 Y 995

Panama 3600 Y 730

Peru 1500 Y 1095

Philippines 2910 Y 905

Poland 350000 N 365

Portugal 350000 N 365

Qatar 0 Y 1095

Romania 350000 N 365

Russian Federation 10000 Y 1095

Saudi Arabia 27000 Y 1095

Senegal 960 Y 1095

Serbia 2500 Y 815

Singapore 69100 Y 730

Slovakia 12000 N 480

Slovenia 350000 N 365

South Africa 7715 Y 1095

Spain 350000 N 365

Sri Lanka 100 Y 605

Sudan 750 Y 1095

Sweden 350000 N 365

Switzerland 60000 N 480

Syria 2000 Y 1095

Taiwan 20000 Y 905

Tajikistan 2050 Y 730

Tanzania 1000 Y 730

Thailand 70 Y 765

Togo 861 Y 1095

100

Country / Region Cost

(USD)

CPP Time for approval

(days)

Trinidad & Tobago 125 Y 545

Turkey 8000 Y 605

Turkmenistan 1400 Y 730

Uganda 1000 Y 730

Ukraine 11000 Y 1095

United Arab Emirates 300 Y 605

United Kingdom 400000 N 365

United States 1958800 N 365

Uzbekistan 5500 Y 905

Venezuela 5232 Y 1095

Vietnam 1000 Y 545

Yemen 700 Y 1095

Zambia 1000 Y 730

Zimbabwe 1000 Y 730

101

Appendix II: Examples of the regulatory process for selected countries119

119

The diagrams are from the Thomson Reuters IDRAC files. The intent of showing the examples in this

section is to illustrate the similarities and differences between the countries. For example, some countries

identify how much time is needed from stage to stage, while others do not. Some countries identify the

different segments or milestones, others do not. Some countries have an appeal process, others do not.

102

Australia

There are eight major milestones or phases in the approval process that is designed to

take up to 13 – 14 months.

Phase I Phase II Phase III Phase IV

Phase V Phase VI Phase VII Phase VIII

103

Chile

There are two major sections of the review process – The pharmaceutical and the

analytical evaluation sections.

104

China

105

Colombia

106

Croatia

Czech Republic

107

Egypt

Estonia

108

EU – Centralized Procedure

109

EU – Mutual Recognition Procedure / Decentralized Process

110

France

111

Indonesia

Israel

112

Italy

113

Japan

Lebanon

114

Netherlands

115

Norway

116

Peru

117

Phillipines

118

Saudi Arabia

119

Singapore

120

Slovakia

121

South Korea

Taiwan

122

United Kingdom

123

Ukraine

124

USA

Serbia

125

Venezuela

126

Appendix III: Listing of countries with matching website for the Drug Health

Authority

127

No Country Link to website of the health authority

1 Algeria http://www.sante.gov.dz/

2 Armenia

http://www.moh.am/?lang=en

http://www.pharm.am/index.php?langid=3

3 Australia www.tga.gov.au

4 Austria http://www.basg.gv.at

5 Azerbaijan http://www.health.gov.az http://www.pharma.az

6 Belarus http://minzdrav.gov.by/ru

7 Bosnia and

Herzegovina http://www.almbih.gov.ba/

8 Botswana http://www.moh.gov.bw/

9 Brazil www.anvisa.gov.br

10 Costa

Rica www.ministeriodesalud.go.cr

11 Croatia http://www.almp.hr/

12 Cyprus www.moh.gov.cy/phs

13 Denmark http://laegemiddelstyrelsen.dk/

14 Dominican

Republic www.drogasyfarmacias.gov.do

15 Ecuador www.inh.gob.ec

16 El Salvador www.salud.gob.sv

17 Ethiopia www.fmhaca.gov.et

18 Finland http://www.fimea.fi/frontpage

19 France http://ansm.sante.fr/

20 Georgia http://www.moh.gov.ge

21 Germany http://www.bfarm.de/DE/Home/home_node.html

22 Ghana www.fdbghana.gov.gh/

23 Guatemala www.mspas.gob.gt

24 Honduras www.salud.gob.hn

25 Hong

Kong http://www.drugoffice.gov.hk

26 Hungary http://www.ogyi.hu/uj_beadvanyok/

27 Iceland http://www.lyfjastofnun.is/

28 Indonesia http://www.pom.go.id/

29 Ireland http://www.imb.ie/

30 Israel http://www.health.gov.il/UnitsOffice/HD/MTI/Drugs/Pages/default.aspx

31 Italy www.agenziafarmaco.it

32 Jamaica www.moh.gov.jm

33 Japan http://www.pmda.go.jp/english/index.html

34 Jordan http://www.jfda.jo/

35 Kazakhstan http://www.dari.kz

36 Kenya www.pharmacyboardkenya.org/

37 Kyrgyzstan

http://www.med.kg

http://www.pharm.kg

38 Lebanon http://www.moph.gov.lb/Drugs/Pages/Drugs.aspx

128

No Country Link to website of the health authority

39 Malawi www.pmpb.mw

40 Malaysia http://www.bpfk.gov.my

41 Malta www.medicinesauthority.gov.mt

42 Mexico www.cofepris.gob.mx

43 Moldova http://www.ms.gov.md/

44 Mongolia http://english.moh.mn/

45 Montenegro http://calims.me

46 Namibia www.nmrc.com.na

47 Nicaragua www.minsa.gob.ni

48 Nigeria www.nafdac.gov.ng

49 Panama www.minsa.gob.pa

50 Philippines http://www.fda.gov.ph/

51 Russian

Federation http://rosminzdrav.ru/docs/mzsr/regulation/1

52 Saudi

Arabia www.sfda.gov.sa

53 Serbia http://www.alims.gov.rs/index_eng.php

54 Singapore

http://www.hsa.gov.sg/publish/hsaportal/en/health_products_regulation/

western_medicines/licences/Fees1.html

55 Slovakia http://www.sukl.sk/en

56 South

Africa www.mccza.com

57 Switzerland www.swissmedic.ch

58 Syria

http://www.moh.gov.sy/ar/%D8%A7%D9%84%D8%B1%D8%A6%D9

%8A%D8%B3%D9%8A%D8%A9/tabid/56/Default.aspx

59 Tajikistan www.health.tj and http://www.pharmnadzor.tj

60 Tanzania www.tfda.or.tz/

61 Thailand http://drug.fda.moph.go.th/drug/

62 Trinidad

&Tobago www.health.gov.tt

63 Uganda www.nda.org.ug

64 Ukraine http://www.pharma-center.kiev.ua/view/

65 United

Kingdom http://www.mhra.gov.uk/Aboutus/Whoweare/index.htm

66 United

States

http://www.fda.gov/ForIndustry/UserFees/PrescriptionDrugUserFee/defa

ult.htm

67 Uzbekistan http://www.minzdrav.uz

68 Venezuela http://www.inhrr.gob.ve/

69 Zambia www.moh.gov.zm

70 Zimbabwe www.mcaz.co.za

129

Appendix IV: Changes in the US drug regulation over time120

120

Source: www.fda.gov

130

Changes in the US drug regulation over time

Date Event

1820 Eleven physicians meet in Washington, D.C., to establish the U.S.

Pharmacopeia, the first compendium of standard drugs for the United States.

1883 Dr. Harvey W. Wiley becomes chief chemist, expanding the Bureau of

Chemistry's food adulteration studies. Campaigning for a federal law, Dr.

Wiley is called the "Crusading Chemist" and "Father of the Pure Food and

Drugs Act." He retired from government service in 1912 and died in 1930.

1898 Association of Official Agricultural Chemists (now AOAC International)

establishes a Committee on Food Standards headed by Dr. Wiley. States

begin incorporating these standards into their food statutes.

1902 The Biologics Control Act is passed to ensure purity and safety of serums,

vaccines, and similar products used to prevent or treat diseases in humans.

Congress appropriates $5,000 to the Bureau of Chemistry to study chemical

preservatives and colors and their effects on digestion and health. Dr. Wiley's

studies draw widespread attention to the problem of food adulteration. Public

support for passage of a federal food and drug law grows.

1906 The original Food and Drugs Act is passed by Congress on June 30 and

signed by President Theodore Roosevelt. It prohibits interstate commerce in

misbranded and adulterated foods, drinks and drugs.

The Meat Inspection Act is passed the same day.

Shocking disclosures of insanitary conditions in meat-packing plants, the use

of poisonous preservatives and dyes in foods, and cure-all claims for

worthless and dangerous patent medicines were the major problems leading

to the enactment of these laws.

1911 In U.S. v. Johnson, the Supreme Court rules that the 1906 Food and Drugs

Act does not prohibit false therapeutic claims but only false and misleading

statements about the ingredients or identity of a drug.

1912 Congress enacts the Sherley Amendment to overcome the ruling in U.S. v.

Johnson. It prohibits labeling medicines with false therapeutic claims

intended to defraud the purchaser, a standard difficult to prove.

131

Changes in the US drug regulation over time

Date Event

1913 Gould Amendment requires that food package contents be "plainly and

conspicuously marked on the outside of the package in terms of weight,

measure, or numerical count."

1914 In U.S. v. Lexington Mill and Elevator Company, the Supreme Court issues

its first ruling on food additives. It ruled that in order for bleached flour with

nitrite residues to be banned from foods, the government must show a

relationship between the chemical additive and the harm it allegedly caused

in humans. The court also noted that the mere presence of such an ingredient

was not sufficient to render the food illegal. The Harrison Narcotic Act

requires prescriptions for products exceeding the allowable limit of narcotics

and mandates increased record-keeping for physicians and pharmacists who

dispense narcotics.

1927 The Bureau of Chemistry is reorganized into two separate entities.

Regulatory functions are located in the Food, Drug, and Insecticide

Administration, and non-regulatory research is located in the Bureau of

Chemistry and Soils.

1930 McNary-Mapes Amendment authorizes FDA standards of quality and fill-of-

container for canned food, excluding meat and milk products.

The name of the Food, Drug, and Insecticide Administration is shortened to

Food and Drug Administration (FDA) under an agricultural appropriations

act.

1933 FDA recommends a complete revision of the obsolete 1906 Food and Drugs

Act. The first bill is introduced into the Senate, launching a five-year

legislative battle.

1938 Elixir of Sulfanilamide, containing the poisonous solvent diethylene glycol,

kills 107 persons, many of whom are children, dramatizing the need to

establish drug safety before marketing and to enact the pending food and

drug law.

1937 The Federal Food, Drug, and Cosmetic (FDC) Act of 1938 is passed by

Congress, containing new provisions:

Extending control to cosmetics and therapeutic devices.

Requiring new drugs to be shown safe before marketing-starting a

132

Changes in the US drug regulation over time

Date Event

new system of drug regulation.

Eliminating the Sherley Amendment requirement to prove intent to

defraud in drug misbranding cases.

Providing that safe tolerances be set for unavoidable poisonous

substances.

Authorizing standards of identity, quality, and fill-of-container for

foods.

Authorizing factory inspections.

Adding the remedy of court injunctions to the previous penalties of

seizures and prosecutions.

Under the Wheeler-Lea Act, the Federal Trade Commission is charged with

overseeing advertising associated with products otherwise regulated by FDA,

with the exception of prescription drugs.

1939 First Food Standards issued (canned tomatoes, tomato purée, and tomato

paste).

1940 FDA transferred from the Department of Agriculture to the Federal Security

Agency, with Walter G. Campbell appointed as the first Commissioner of

Food and Drugs.

1941- Insulin Amendment requires FDA to test and certify purity and

potency of this lifesaving drug for diabetes.

1943 In U.S. v. Dotterweich, the Supreme Court rules that the responsible officials

of a corporation, as well as the corporation itself, may be prosecuted for

violations. It need not be proven that the officials intended, or even knew of,

the violations.

1945 Penicillin Amendment requires FDA testing and certification of safety and

effectiveness of all penicillin products. Later amendments extended this

requirement to all antibiotics. In 1983 such control was found no longer

needed and was abolished.

1949 FDA publishes guidance to industry for the first time. This guidance,

"Procedures for the Appraisal of the Toxicity of Chemicals in Food," came to

be known as the "black book."

133

Changes in the US drug regulation over time

Date Event

1950 In Alberty Food Products Co. v. U.S., a court of appeals rules that the

directions for use on a drug label must include the purpose for which the drug

is offered. Therefore, a worthless remedy cannot escape the law by not

stating the condition it is supposed to treat.

Oleomargarine Act requires prominent labeling of colored oleomargarine, to

distinguish it from butter.

Delaney Committee starts congressional investigation of the safety of

chemicals in foods and cosmetics, laying the foundation for the 1954 Miller

Pesticide Amendment, the 1958 Food Additives Amendment, and the 1960

Color Additive Amendment.

1951 Durham-Humphrey Amendment defines the kinds of drugs that cannot be

safely used without medical supervision and restricts their sale to prescription

by a licensed practitioner.

1952 In U.S. v. Cardiff, the Supreme Court rules that the factory inspection

provision of the 1938 FDC Act is too vague to be enforced as criminal law.

FDA consumer consultants are appointed in each field district to maintain

communications with consumers and ensure that FDA considers their needs

and problems.

1954 Miller Pesticide Amendment spells out procedures for setting safety limits for

pesticide residues on raw agricultural commodities.

First large-scale radiological examination of food carried out by FDA when it

received reports that tuna suspected of being radioactive was being imported

from Japan following atomic blasts in the Pacific. FDA begins monitoring

around the clock to meet the emergency.

1955 HEW Secretary Oveta Culp Hobby appoints a committee of 14 citizens to

study the adequacy of FDA's facilities and programs. The committee

recommends a substantial expansion of FDA staff and facilities, a new

headquarters building, and more use of educational and informational

programs.

1962 Thalidomide, a new sleeping pill, is found to have caused birth defects in

thousands of babies born in Western Europe. News reports on the role of Dr.

Frances Kelsey, FDA medical officer, in keeping the drug off the U.S.

134

Changes in the US drug regulation over time

Date Event

market, arouse public support for stronger drug regulation.

Kefauver-Harris Drug Amendments passed to ensure drug efficacy and

greater drug safety. For the first time, drug manufacturers are required to

prove to FDA the effectiveness of their products before marketing them.

Consumer Bill of Rights is proclaimed by President John F. Kennedy in a

message to Congress. Included are the right to safety, the right to be

informed, the right to choose, and the right to be heard.

1965 Drug Abuse Control Amendments are enacted to deal with problems caused

by abuse of depressants, stimulants and hallucinogens.

1966 FDA contracts with the National Academy of Sciences/National Research

Council to evaluate the effectiveness of 4,000 drugs approved on the basis of

safety alone between 1938 and 1962.

Child Protection Act enlarges the scope of the Federal Hazardous Substances

Labeling Act to ban hazardous toys and other articles so hazardous that

adequate label warnings could not be written.

Fair Packaging and Labeling Act requires all consumer products in interstate

commerce to be honestly and informatively labeled, with FDA enforcing

provisions on foods, drugs, cosmetics, and medical devices.

1968 FDA Bureau of Drug Abuse Control and Treasury Department Bureau of

Narcotics are transferred to the Department of Justice to form the Bureau of

Narcotics and Dangerous Drugs (BNDD), consolidating efforts to police

traffic in abused drugs.

Reorganization of federal health programs places FDA in the Public Health

Service.

FDA forms the Drug Efficacy Study Implementation (DESI) to implement

recommendations of the National Academy of Sciences investigation of

effectiveness of drugs first marketed between 1938 and 1962.

1970 In Upjohn v. Finch the Court of Appeals upholds enforcement of the 1962

drug effectiveness amendments by ruling that commercial success alone does

not constitute substantial evidence of drug safety and efficacy.

FDA requires the first patient package insert: oral contraceptives must

contain information for the patient about specific risks and benefits.

The Comprehensive Drug Abuse Prevention and Control Act replaces

135

Changes in the US drug regulation over time

Date Event

previous laws and categorizes drugs based on abuse and addiction potential

compared to their therapeutic value.

Environmental Protection Agency established; takes over FDA program for

setting pesticide tolerances.

1972 Over-the-Counter Drug Review begun to enhance the safety, effectiveness

and appropriate labeling of drugs sold without prescription.

1973 The U.S. Supreme Court upholds the 1962 drug effectiveness law and

endorses FDA action to control entire classes of products by regulations

rather than to rely only on time-consuming litigation.

Consumer Product Safety Commission created by Congress; takes over

programs pioneered by FDA under 1927 Caustic Poison Act, 1960 Federal

Hazardous Substances Labeling Act, 1966 Child Protection Act, and PHS

accident prevention activities for safety of toys, home appliances, etc.

1976 Medical Device Amendments passed to ensure safety and effectiveness of

medical devices, including diagnostic products. The amendments require

manufacturers to register with FDA and follow quality control procedures.

Some products must have pre-market approval by FDA; others must meet

performance standards before marketing.

1977 Saccharin Study and Labeling Act passed by Congress to stop FDA from

banning the chemical sweetener but requiring a label warning that it has been

found to cause cancer in laboratory animals.

Introduction of the Bioresearch Monitoring Program as an agency-wide

initiative ensures the quality and integrity of data submitted to FDA and

provides for the protection of human subjects in clinical trials by focusing on

preclinical studies on animals, clinical investigations, and the work of

institutional review boards.

1979 In the hours following the Three Mile Island nuclear emergency of March 28,

1979, FDA contracted with firms in Missouri, Michigan, and New Jersey to

prepare and package enough doses of potassium iodide to protect those

threatened with thyroid cancer if exposed to radiation. Nearly one quarter of a

million bottles-enough for every household in the area-were delivered to

Harrisburg, Pennsylvania within 72 hours.

136

Changes in the US drug regulation over time

Date Event

1980 Infant Formula Act establishes special FDA controls to ensure necessary

nutritional content and safety.

1982 Tamper-resistant Packing Regulations issued by FDA to prevent poisonings

such as deaths from cyanide placed in Tylenol capsules. The Federal Anti-

Tampering Act passed in 1983 makes it a crime to tamper with packaged

consumer products.

FDA publishes first Red Book (successor to 1949 "black book"), officially

known as Toxicological Principles for the Safety Assessment of Direct Food

Additives and Color Additives Used in Food.

1983 Orphan Drug Act passed, enabling FDA to promote research and marketing

of drugs needed for treating rare diseases.

1984 Fines Enhancement Laws of 1984 and 1987 amend the U.S. Code to greatly

increase penalties for all federal offenses. The maximum fine for individuals

is now $100,000 for each offense and $250,000 if the violation is a felony or

causes death. For corporations, the amounts are doubled.

1985 AIDS test for blood approved by FDA in its first major action to protect

patients from infected donors.

1986 Childhood Vaccine Act requires patient information on vaccines, gives FDA

authority to recall biologics, and authorizes civil penalties.

1987 Investigational drug regulations revised to expand access to experimental

drugs for patients with serious diseases with no alternative therapies.

1988 Food and Drug Administration Act of 1988 officially establishes FDA as an

agency of the Department of Health and Human Services with a

Commissioner of Food and Drugs appointed by the President with the advice

and consent of the Senate, and broadly spells out the responsibilities of the

Secretary and the Commissioner for research, enforcement, education, and

information.

1989 FDA issues a nationwide recall of all over-the-counter dietary supplements

containing 100 milligrams or more of L-Tryptophan, due to a clear link

137

Changes in the US drug regulation over time

Date Event

between the consumption of L-tryptophan tablets and its association with a

U.S. outbreak of Eosinophilia Myalgia Syndrome (EMS), characterized by

fatigue, shortness of breath, and other symptoms. By 1990 the Centers for

Disease Control and Prevention confirm over 1,500 cases of EMS, including

38 deaths, and FDA prohibits the importation of l-tryptophan.

1991 Regulations published to Accelerate the Review of Drugs for life-threatening

diseases.

1992 Generic Drug Enforcement Act imposes debarment and other penalties for

illegal acts involving abbreviated drug applications.

Prescription Drug User Fee Act (PDUFA) requires drug and biologics

manufacturers to pay fees for product applications and supplements, and

other services. The act also requires FDA to use these funds to hire more

reviewers to assess applications.

1993 A consolidation of several adverse reaction reporting systems is launched as

MedWatch, designed for voluntary reporting of problems associated with

medical products to be filed with FDA by health professionals.

1995 FDA declares cigarettes to be "drug delivery devices." Restrictions are

proposed on marketing and sales to reduce smoking by young people.

1996 Federal Tea Tasters Repeal Act repeals the Tea Importation Act of 1897 to

eliminate the Board of Tea Experts and user fees for FDA's testing of all

imported tea. Tea itself is still regulated by FDA.

1997 Food and Drug Administration Modernization Act reauthorizes the

Prescription Drug User Fee Act of 1992 and mandates the most wide-ranging

reforms in agency practices since 1938. Provisions include measures to

accelerate review of devices, regulate advertising of unapproved uses of

approved drugs and devices, and regulate health claims for foods.

1999 ClinicalTrials.gov is founded to provide the public with updated information

on enrollment in federally and privately supported clinical research, thereby

expanding patient access to studies of promising therapies.

A final rule mandates that all over-the-counter drug labels must contain data

in a standardized format. These drug facts are designed to provide the patient

138

Changes in the US drug regulation over time

Date Event

with easy-to-find information, analogous to the nutrition facts label for foods.

2000 The U. S. Supreme Court, upholding an earlier decision in Food and Drug

Administration v. Brown & Williamson Tobacco Corp. et al., ruled 5-4 that

FDA does not have authority to regulate tobacco as a drug. Within weeks of

this ruling, FDA revokes its final rule, issued in 1996, that restricted the sale

and distribution of cigarettes and smokeless tobacco products to children and

adolescents, and that determined that cigarettes and smokeless tobacco

products are combination products consisting of a drug (nicotine) and device

components intended to deliver nicotine to the body.

Federal agencies are required to issue guidelines to maximize the quality,

objectivity, utility, and integrity of the information they generate, and to

provide a mechanism whereby those affected can secure correction of

information that does not meet these guidelines, under the Data Quality Act.

2003 The Medicare Prescription Drug Improvement and Modernization Act

requires, among other elements, that a study be made of how current and

emerging technologies can be utilized to make essential information about

prescription drugs available to the blind and visually impaired.

To help consumers choose heart-healthy foods, the Department of Health and

Human Services announces that FDA will require food labels to include

trans-fat content, the first substantive change to the nutrition facts panel on

foods since the label was changed in 1993. An obesity working group is

established by the Commissioner of Food and Drugs, charged to develop an

action plan to deal with the nation's obesity epidemic from the perspective of

FDA. In March 2004 the group releases "Calories Count: Report of the

Obesity Working Group," which addresses issues connected to the food label,

obesity therapeutics, research needs, the role of education, and other topics.

2004 Project BioShield Act of 2004 authorizes FDA to expedite its review

procedures to enable rapid distribution of treatments as countermeasures to

chemical, biological, and nuclear agents that may be used in a terrorist attack

against the U. S., among other provisions.

Passage of the Food Allergy Labeling and Consumer Protection Act requires

the labeling of any food that contains a protein derived from any one of the

following foods that, as a group, account for the vast majority of food

allergies: peanuts, soybeans, cow's milk, eggs, fish, crustacean shellfish, tree

nuts, and wheat.

139

Changes in the US drug regulation over time

Date Event

A ban on over-the-counter steroid precursors, increased penalties for making,

selling, or possessing illegal steroids precursors, and funds for preventive

education to children are features of the Anabolic Steroid Control Act of

2004.

FDA publishes "Innovation or Stagnation? -- Challenge and Opportunity on

the Critical Path to New Medical Products," which examines the critical path

needed to bring therapeutic products to fruition, and how FDA can

collaborate in the process, from laboratory to production to end use, to make

medical breakthroughs available to those in need as quickly as possible.

2005 Formation of the Drug Safety Board is announced, consisting of FDA staff

and representatives from the National Institutes of Health and the Veterans

Administration. The Board will advise the Director, Center for Drug

Evaluation and Research, FDA, on drug safety issues and work with the

agency in communicating safety information to health professionals and

patients

140

Appendix V: NDA required sections in the United States

141

The New Drug Application (NDA)

In the US, pharmaceuticals fall under the Food, Drug, and Cosmetic Act (FD&C Act)

which contains 10 chapters and significant amendments totaling hundreds of pages. The

FD&C is part of the Code of Federal Register (CFR). There are 50 titles in the CFR and

title 21 refers to Food and Drugs totaling 4436 pages of regulation!

Below is the listing of NDA sections121

:

1. Index

2. Labeling

3. Summary (21 CFR 314.50 (c) )

4. Chemistry Section

a. Chemistry, Manufacturing and Controls (CMC)

b. Samples (21 CFR 314.50 (e)(1); 21 CFR 601.2 (a)) (Submit only upon FDA’s

request)

c. Methods validation package (e.g., 21 CFR 314.50(e)(2)(i); 21 CFR 601.2)

5. Nonclinical pharmacology and toxicology section (e.g., 21 CFR 314.50(d)(2); 21 CFR

601.2)

6. Human pharmacokinetics and bioavailability section (e.g., 21 CFR 314.50(d)(3); 21

CFR 601.2)

7. Clinical microbiology section (e.g., 21 CFR 314.50(d)(4))

8. Clinical data section (e.g., 21 CFR 314.50(d)(5); 21 CFR 601.2)

9. Safety update report (e.g., 21 CFR 314.50(d)(5)(vi)(b);21 CFR 601.2)

10. Statistical section (e.g., 21 CFR 314.50(d)(6); 21 CFR 601.2)

11. Case report tabulations (e.g., 21 CFR 314.50(f)(1);21 CFR 601.2)

12. Case report forms (e.g., 21 CFR 314.50 (f)(2); 21 CFR 601.2)

13. Patent information on any patent that claims the drug/biologic (21 U.S.C. 355(b) or

(c))

14. A patent certification with respect to any patent that claims the drug/biologic (21

U.S.C. 355 (b)(2) or (j)(2)(A))

15. Establishment description (21 CFR Part 600, if applicable)

16. Debarment certification (FD&C Act 306 (k)(1))

17. Field copy certification (21 CFR 314.50 (l)(3))

18. User Fee Cover Sheet (PDUFA Form FDA 3397, GDUFA Form FDA 3794, BsUFA

Form FDA 3792, or MDUFMA Form FDA 3601)

19. Financial Disclosure Information (21 CFR Part 54)

121

Information retrieved from www.fda.gov

http://www.fda.gov/drugs/developmentapprovalprocess/formssubmissionrequirements/drugmasterfilesdmfs

/ucm073080.htm

142

Appendix VI: Regression analysis data with Time as the dependent variable

143

TABLE 6

RESULTS OF REGRESSION ANALYSIS OF APPROVAL TIME ON COUNTRY ECONOMIC AND POLITICAL

CHARACTERISTICS ACROSS 129 COUNTRIES, 2010 - 2011

Variables OLS

(t-Stat) (1) (2) (3) (4) (5) (6) (7)

LG GDP PC -47.50 -34.88 -4.01 -6.33

(-3.04)* (-2.28)* (-0.26) (-0.42)

Cost -0.00044 -7.42E-05 -7.40E-05 -0.00013 -0.00013

(-3.59)* (0.00013) (-0.58) (-0.99) (-0.99)

Health Expend -0.081 -0.087 -0.082 -0.074 -0.076

(-5.57)* (-7.80)* (-6.20)* (-4.94)* (-5.58)*

Open K -0.72 -0.72

(-1.86) (-1.88)

Constant 1118 1044 875 845 844 954 905

(8.84) (8.52) (7.47) (33.55) (33.39) (7.73) (22.08)

Adjusted R2

0.063 0.15 0.32 0.33 0.32 0.33 0.34

# of observations 124 124 121 125 125 121 125

*statistically significant

144

TABLE 6 (continued)

Variables OLS

(t-Stat) (8) (9) (10) (11) (12) (13) (14)

LG GDP PC 3.38 12.64

(0.23) (0.76)

Cost -0.00013 -0.00013 -0.00012 -0.00013 -0.00013 -0.00012 -0.00015

(-0.97) (-1.10) (-1.05) (-1.07) (-0.97) (-0.90) (-1.35)

Health Expend -0.075 -0.056 -0.056 -0.040 -0.052 -0.055 -0.035

(-5.44)* (-3.97)* (3.80)* (-2.46)* (-3.06)* (-3.13)* (-2.26)*

Open K -0.72 -0.43 -0.39 -0.13 -0.33 -0.25 -0.044

(-1.87) (-1.22) (-1.07) (-0.33) (-0.79) (-0.59) (-0.12)

Dem Score -0.44

(-0.21)

Phys / 1000 -49.84 -50.52 -28.45

(-2.99)* (-2.90)* (-1.50)

Frag. Index 12.15 13.01 12.64 7.16

(2.29)* (2.78) (2.71)* (1.40)

Life Expectancy -7.93

(-2.47)*

Constant 906 934 902 755 738 623 1296

(21.69) (21.08) (7.48) (8.77) (10.24) (3.67) (5.19)

Adjusted R2

0.33 0.42 0.41 0.42 0.35 0.35 0.41

# of observations 125 106 103 103 121 117 121

145

TABLE 6 (continued)

Variables OLS

(t-Stat) (15) (16) (17) (18)

LG GDPPC 11.56

(0.73)

Cost -0.00015 -0.00027 -0.00028 -0.00015

(-1.33) (-2.56) (-2.68)* (-1.27)

Health Expend -0.031 -0.036

(-1.89) (-2.17)*

Open K -0.020 0.144 0.018 -0.057

(-0.052) (0.36) (0.048) (-0.15)

KG 0.96 0.27 1.50

(0.21) (0.060) (0.34)

Phys / 1000 -19.21 -37.08 -37.15 -26.36

(-0.97) (-1.96) (-1.98) (-1.38)

Frag. Index 9.60 19.26 17.07 12.48

(1.64) (3.48)* (3.41)* (2.34)*

Life Expectancy -4.75

(-1.23)

Constant 1077 549 681 727

(3.74) (3.06) (7.29) (7.73)

Adjusted R2

0.41 0.38 0.38 0.40

# of observations 103 100 103 103

*statistically significant

146

TABLE 6 (continued)

Variables OLS

(t-Stat) (19) (20) (21) (22) (23) (24) (25)

GDP PC -1.55e-11 -1.60e-11 1.97e-11

(-1.08) (-1.12) (0.89)

Cost -0.00038

(-2.09)*

Open K 0.077

(0.17)

Frag. Index 21.16 20.90 13.30 12.97

(5.07)* (5.43)* (2.29)* (2.23)*

Life Expectancy -7.061 6.55 -13.67 -12.92

(-1.89) (-1.74) (-5.68)* (-5.38)*

Democracy 0.162 -2.67

(0.07) (-1.09)

Constant 512 522 699 1074 1049 1650 1600

(7.78) (13.88) (24.86) (3.59) (3.50) (9.77) (9.51)

Adjusted R2

0.17 0.19 0.0014 0.22 0.22 0.22 0.24

# of observations 124 124 128 123 123 128 128

*statistically significant

147

TABLE 6 (continued)

Variables OLS

(t-Stat) (26) (27) (28) (29) (30) (31) (32) (33)

LG GDP PC -3.5e-11

(-2.27)*

Cost -0.00029 -0.00024 -0.00020 0.00093

(-2.46)* (-2.03)* (-1.74) (-3.06)*

Literacy rate -5.37 -2.033 -4.032

(-3.16)* (-0.85) (-2.42)*

Phys / 1000 -74.85 -65.29 -31.23 -15.72

(-4.32)* (-3.76)* (-1.37) (-0.73)

Frag. Index 18.80

(3.35)*

Life Expectancy -8.62 -8.31

(-2.25)* (-1.93)*

Constant 799 800 1343 557 702 1175 1459 1094

(18.74) (19.22) (5.49) (6.86) (26.74) (8.01) (7.09) (7.75)

Adjusted R2

0.144 0.18 0.21 0.25 0.031 0.11 0.14 0.20

# of observations 106 106 106 103 129 73 73 73

*statistically significant

148

Curriculum Vitae

SUMMARY

A technically oriented, experienced, dedicated team leader with extensive knowledge & experience in the drug development process [from discovery through launch] and exceptional expertise in clinical trial supply management. Proven track [12 years] of successful strategy development and project execution through all stages of drug development, across continents and cultures building lasting bridges along the way. Hands on experience in sterile and solid dosage manufacturing, demand & supply forecasting, and project management, execution & leadership.

EXPERIENCE

Novartis Consumer Health May’13 – Present

Associate Director, Project Management (05/13 – Present)

Product Sustainment and Strategy PMO

Merck & Co. Nov '09 – May’13

Senior Manager, Business Consulting (11/09 – 05/13)

Organizational Strategy Management and Operations, Merck Manufacturing Division

Schering-Plough Corp Nov '01 – Nov'09

Senior Manager, Global Development and Commercialization PMO, Global Supply Chain (1/09 – 11/09)

Project Manager, Global Clinical Supplies Planning Kenilworth, NJ (6/05 – 1/09)

Investigational Material Specialist, Global Clinical Supplies Kenilworth, NJ (11/01 – 6/05) EDUCATION Rutgers University, Newark, NJ 05/14 Doctor of Philosophy, Global Affairs Rutgers University, Newark, NJ 05/10 Master of Science, Global Affairs New Jersey Institute of Technology, Newark, NJ 05/05 Master of Science, Pharmaceutical Engineering New Jersey Institute of Technology, Newark, NJ 05/01 Bachelor of Science, Chemical Engineering INTERNATIONAL EXPERIENCE Oss, Netherlands; Lucerne, Switzerland; Antwerp, Belgium; Cork, Ireland; Mexico City, Mexico Middle East (Jordan, Syria, Lebanon, Egypt), France, Spain, Italy, Germany, England LANGUAGE Fluent in English and Arabic Familiar in French, Dutch & German LinkedIn Profile: http://www.linkedin.com/profile/view?id=28066458&trk=tab_pro


Recommended