You are on page 1of 17

Chapter 20 

Regulatory affairs
I Hägglöf, Å Holmgren

uses. These developed into official pharmacopoeias, of


INTRODUCTION which the earliest was probably the New Compound Dis-
pensatory of 1498 issued by the Florentine guild of physi-
This chapter introduces the reader to the role of the regula- cians and pharmacists.
tory affairs (RA) department of a pharmaceutical company, The pharmacopoeias were local rules, applicable in a
outlining the process of getting a drug approved, and particular city or district. During the 19th century national
emphasizing the importance of interactions of regulatory pharmacopoeias replaced local ones, and since the early
affairs with other functions within the company, and with 1960s regional pharmacopoeias have successively replaced
the external regulatory authorities. national ones. Now work is ongoing to harmonize – or
To keep this chapter to a reasonable size the typical at least mutually recognize – interchangeable use of the
examples given refer to the first registration of a new chem- US Pharmacopeia, the European Pharmacopoeia and the
ical compound. The same way of reasoning also applies, Japanese Pharmacopoeia.
however, to any subsequent change to the approval of As described in Chapter 1, the development of experi-
products. Depending on the magnitude of the change, the mental pharmacology and chemistry began during the
new documentation that needs to be compiled, submitted second half of the 19th century, revealing that the effect of
and approved by health authorities is variable, ranging the main botanical drugs was due to chemical substances
from a few pages of pharmaceutical data (e.g. for an in the plants used. The next step, synthetic chemistry, made
update to product stability information) to a complete it possible to produce active chemical compounds. Other
new application for a new clinical use in a new patient important scientific developments, e.g. biochemistry, bac-
group in a new pharmaceutical form. teriology and serology during the early 20th century, accel-
It needs also to be said that, as every drug substance and erated the development of the pharmaceutical industry
every project is unique, the views expressed represent the into what it is today (see Drews, 1999).
opinion of the authors and are not necessarily shared by Lack of adequate drug control systems or methods to
others active in the field. investigate the safety of new chemical compounds became
a great risk as prefabricated drug products were broadly
and freely distributed. In the USA the fight against patent
medicines led to the passing of the US Pure Food and
BRIEF HISTORY OF
Drugs Act against misbranding as long ago as 1906. The
PHARMACEUTICAL REGULATION Act required improved declaration of contents, prohibited
false or misleading statements, and required content and
Control of pharmaceutical products has been the task of purity to comply with labelled information. A couple of
authorized institutions for thousands of years, and this decades later, the US Food and Drug Administration
was the case even in ancient Greece and Egypt. (FDA) was established to control US pharmaceutical
From the Middle Ages, control of drug quality, composi- products.
tion purity and quantification was achieved by reference Safety regulations in the USA were, however, not enough
to authoritative lists of drugs, their preparation and their to prevent the sale of a paediatric sulfanilamide elixir

© 2012 Elsevier Ltd. 285


Section | 3 | Drug Development

containing the toxic solvent diethylene glycol. In 1937, The 1960s and 1970s saw a rapid increase in laws, regu-
107 people, both adults and children, died as a result of lations and guidelines for reporting and evaluating the
ingesting the elixir, and in 1938 the Food Drug and Cos- risks versus the benefits of new medicinal products. At the
metics Act was passed, requiring for the first time approval time the industry was becoming more international and
by the FDA before marketing of a new drug product. seeking new global markets, but the registration of medi-
The thalidomide disaster further demonstrated the lack cines remained a national responsibility.
of adequate drug control. Thalidomide (Neurosedyn®, Although different regulatory systems were based on the
Contergan®) was launched during the last years of the same key principles, the detailed technical requirements
1950s as a non-toxic treatment for a variety of conditions, diverged over time, often for traditional rather than scien-
such as colds, anxiety, depression, infections, etc., both tific reasons, to such an extent that industry found it neces-
alone and in combination with a number of other com- sary to duplicate tests in different countries to obtain
pounds, such as analgesics and sedatives. global regulatory approval for new products. This was a
The reason why the compound was regarded as harmless waste of time, money and animals’ lives, and it became
was the lack of acute toxicity after high single doses. After clear that harmonization of regulatory requirements was
repeated long-term administration, however, signs of neu- needed.
ropathy developed, with symptoms of numbness, paraes- European (EEC) efforts to harmonize requirements for
thesia and ataxia. But the overwhelming effects were the drug approval began 1965, and a common European
gross malformation in infants born to mothers who had approach grew with the expansion of the European Union
taken thalidomide in pregnancy: their limbs were partially (EU) to 15 countries, and then 27. The EU harmonization
or totally missing, a previously extremely rare malforma- principles have also been adopted by Norway and Iceland.
tion called phocomelia (seal limb). Altogether around This successful European harmonization process gave
12 000 infants were born with the defect in those few years. impetus to discussions about harmonization on a broader
Thalidomide was withdrawn from the market in 1961/62. international scale (Cartwright and Matthews, 1994).
This catastrophe became a strong driver to develop
animal test methods to assess drug safety before testing
compounds in humans. Also it forced national authorities
to strengthen the requirements for control procedures INTERNATIONAL HARMONIZATION
before marketing of pharmaceutical products (Cartwright
and Matthews, 1991). The harmonization process started in 1990, when repre-
Another blow hit Japan between 1959 and 1971. The sentatives of the regulatory authorities and industry asso-
SMON (subacute myelo-optical neuropathy) disaster was ciations of Europe, Japan and the USA (representing the
blamed on the frequent Japanese use of the intestinal majority of the global pharmaceutical industry) met,
antiseptic clioquinol (Entero-Vioform®, Enteroform® or ostensibly to plan an International Conference on Harmo-
Vioform®). The product had been sold without restrictions nization (ICH). The meeting actually went much further,
since early 1900, and it was assumed that it would not be suggesting terms of reference for ICH, and setting up an ICH
absorbed, but after repeated use neurological symptoms Steering Committee representing the three regions.
appeared, characterized by paraesthesia, numbness and The task of ICH was ‘… increased international harmo-
weakness of the extremities, and even blindness. SMON nization, aimed at ensuring that good quality, safe and
affected about 10 000 Japanese, compared to some 100 effective medicines are developed and registered in the
cases in the rest of the world (Meade, 1975). most efficient and cost-effective manner. These activities
These tragedies had a strong impact on governmental are pursued in the interest of the consumer and public
regulatory control of pharmaceutical products. In 1962 health, to prevent unnecessary duplication of clinical trials
the FDA required evidence of efficacy as well as safety as in humans and to minimize the use of animal testing
a condition for registration, and formal approval was without compromising the regulatory obligations of safety
required for patients to be included in clinical trials of and effectiveness’ (Tokyo, October 1990).
new drugs. ICH has remained a very active organization, with
In Europe, the UK Medicines Act 1968 made safety substantial representation at both authority and industry
assessment of new drug products compulsory. The Swedish level from the EU, the USA and Japan. The input of other
Drug Ordinance of 1962 defined the medicinal product nations is provided through World Health Organization
and required a clear benefit–risk ratio to be documented representatives, as well as representatives from Switzerland
before approval for marketing. All European countries and Canada.
established similar controls during the 1960s. ICH conferences, held every 2 years, have become a
In Japan, the Pharmaceutical Affairs Law enacted in 1943 forum for open discussion and follow-up of the topics
was revised in 1961,1979 and 2005 to establish the current decided. The important achievements so far are the scien-
drug regulatory system, with the Ministry of Health and tific guidelines agreed and implemented in the national/
Welfare assessing drugs for quality, safety and efficacy. regional drug legislation, not only in the ICH territories

286
Regulatory affairs Chapter | 20 |

The regulatory authority:

Step 5 Implementation in the three regions • approves clinical trial applications


• gives procedural and scientific advice to companies
during drug development
Step 4 Agreement on a harmonized ICH guideline
Adopted by regulators
• approves for marketing drugs that have been
scientifically evaluated to provide evidence of a
Step 3 Regulatory consultation in the three regions satisfactory benefit/risk ratio
Consolidation of the comments • monitors the safety of the marketed product, based
on (a) reports of adverse reactions from healthcare
Step 2 Agreement by the Steering Committee to release providers, and (b) from compiled and evaluated
the draft consensus text for wider consultation safety information from the company that owns the
Step 1 Building scientific concensus in joint Regulatory/Industry product
Expert Working Groups • can withdraw the licence for marketing in serious
cases of non-compliance (e.g. failure on inspections,
failure of adequate additional warnings in
Fig. 20.1  Five steps in the ICH process for harmonization of prescribing information after clinical adverse
technical issues. reactions are reported, or failure of the company to
consider serious findings in animal studies).
The company:
but also in other countries around the world. So far some • owns the documentation that forms the basis for
50 guidelines have reached ICH approval and regional assessment, is responsible for its accuracy and
implementation, i.e. steps 4 and 5 (Figure 20.1). For a correctness, for keeping it up to date, and for ensuring
complete list of ICH guidelines and their status, see the that it complies with standards set by current scientific
ICH website (website reference 1). development and the regulatory authorities
The process described in Figure 20.1 is very open, and • collects, compiles and evaluates safety data, and
the fact that health authorities and the pharmaceutical submits reports to the regulatory authorities at
industry collaborate from the start increases the efficiency regular intervals – and takes rapid action in serious
of work and ensures mutual understanding across regions cases. This might involve the withdrawal of the entire
and functions; this is a major factor in the success of ICH. product or of a product batch (e.g. tablets containing
the wrong drug or the wrong dose), or a request to
the regulatory authority for a change in prescribing
information
ROLES AND RESPONSIBILITIES  
• has a right to appeal and to correct cases of
OF REGULATORY AUTHORITY   non-compliance.
AND COMPANY
The role of the regulatory  
The basic division of responsibilities for drug products is
affairs department
that the health authority is protecting public health and
safety, and the pharmaceutical company is responsible for The regulatory affairs (RA) department of a pharmaceuti-
all aspects of the drug product. The regulatory approval of cal company is responsible for obtaining approval for new
a pharmaceutical product permits marketing and is a con- pharmaceutical products and ensuring that approval is
tract between the regulatory authority and the pharmaceu- maintained for as long as the company wants to keep the
tical company. The conditions of the approval are set out product on the market. It serves as the interface between
in the dossier and condensed in the prescribing informa- the regulatory authority and the project team, and is the
tion. Any change that is planned must be forwarded to the channel of communication with the regulatory authority
regulatory authority for information and, in most cases, as the project proceeds, aiming to ensure that the project
new approval before being implemented. plan correctly anticipates what the regulatory authority
To protect the public health, regulatory authorities also will require before approving the product. It is the respon-
develop regulations and guidelines for companies to sibility of RA to keep abreast of current legislation, guide-
follow in order to achieve a balance between the possible lines and other regulatory intelligence. Such rules and
risks and therapeutic advantages to patients. The authori- guidelines often allow some flexibility, and the regulatory
ties’ work is partly financed by fees paid by pharmaceutical authorities expect companies to take responsibility for
companies. Fees may be reduced, under certain condi- deciding how they should be interpreted. The RA depart-
tions, to stimulate research. This may be driven, e.g., by ment plays an important role in giving advice to the
company size or size of target patient groups. project team on how best to interpret the rules. During

287
Section | 3 | Drug Development

the development process sound working relations with proposed. In the ICH environment there is a greater pos-
authorities are essential, e.g. to discuss such issues as diver- sibility to exert influence at an early stage.
gence from guidelines, the clinical study programme, and
formulation development.
Most companies assess and prioritize new projects
based on an intended Target Product Profile (TPP). The RA
THE DRUG DEVELOPMENT PROCESS
professional plays a key role in advising on what will be
realistic prescribing information (‘label’) for the intended An overview of the process of drug development is given
product. As a member of the project team RA also contrib- in Chapters 14–18 and summarized in Figure 20.2. As
utes to designing of the development programme. The RA already emphasized, this sequential approach, designed to
department reviews all documentation from a regulatory minimize risk by allowing each study to start only when
perspective, ensuring that it is clear, consistent and com- earlier studies have been successfully completed, is giving
plete, and that its conclusions are explicit. The department way to a partly parallel approach in order to save develop-
also drafts the core prescribing information that is the ment time.
basis for global approval, and will later provide the plat- All studies in the non-clinical area – chemistry, pharma-
form for marketing. The documentation includes clinical cology, pharmacokinetics, pharmaceutical development
trials applications, as well as regulatory submissions for and toxicology – aim to establish indicators of safety and
new products and for changes to approved products. The efficacy sufficient to allow studies and use in man. Accord-
latter is a major task and accounts for about half of the ing to ICH nomenclature, documentation of chemical and
work of the RA department. pharmaceutical development relates to quality assessment,
An important proactive task of the RA is to provide animal studies relate to safety assessment, and studies in
input when legislative changes are being discussed and humans relate to efficacy.

Preclinical studies Clinical studies

Discovery Development

Chemistry/
pharmacology IND* Phase I Phase II Phase III NDA** Phase IV

Search for active Regulatory Efficacy studies Clinical studies Comparative Regulatory Continued
substances view on healthy on a limited scale studies on a review comparative
Toxicology, volunteers 100 – 200 large number studies
efficacy studies 50 –150 patients of patients Registration,
on various persons 500 – 5000 market
types of animals patients introduction
*Investigational
new drug
Application for Knowledge level
permission to
administer a new **New drug
drug to humans application
Application for
Knowledge level permission to
market
a new drug

Time span

2–4 years 2–6 months 3–6 years 1–3 years

10 –13 years from idea to marketable drug

Fig. 20.2  The drug development process.

288
Regulatory affairs Chapter | 20 |

Quality assessment (chemistry and not to create local rules to avoid, as far as possible, differ-
ent interpretations and duplicate work.
pharmaceutical development)
As previously said, all changes to the originally submit-
The quality module of a submission documents purity and ted dossier must be made known to the approving regula-
assay for the drug substance, and purity data for all the tory authority. Since the majority of changes are made in
inactive ingredients. The formulation must fulfil require- the quality section, very small changes take lots of resources
ments for consistent quality and allow storage, and the for the company as well as for the authority. The US leg-
container must be shown to be fit for its purpose. These islation has allowed the submission of annual reports col-
aspects of a pharmaceutical product have to be kept under lecting those changes that have no impact on quality. This
control throughout the development process, as toxicol- possibility has also been introduced in EU in 2010 with
ogy and pharmacology results are reliable only for sub- the purpose of saving time and resources.
stances of comparable purity. Large-scale production,
improved synthetic route, different raw material supply,
etc., may produce a substance somewhat different from Safety assessment (pharmacology
the first laboratory-scale batches. Any substantial change and toxicology)
must be known and documented. Next we consider how to design and integrate pharmaco-
The formulation of a product is a challenge. For initial logical and toxicological studies in order to produce ade-
human studies, simple i.v. and oral solutions are needed quate documentation for the first tests in humans. ICH
for straightforward results, whereas for the clinical pro- guidelines define the information needed from animal
gramme in patients, bioequivalent formulations are essen- studies in terms of doses and time of exposure, to allow
tial for comparison of results across studies, and so it is clinical studies, first in healthy subjects and later in
preferable to have access to the final formulation already patients. The principles and methodology of animal
during Phase II. studies are described in Chapters 11 and 15. The questions
If the formulation intended for marketing cannot be discussed here are when and why these animal studies are
completed until late in the clinical phase, bioequivalence required for regulatory purposes.
studies showing comparable results with the preliminary
and final market formulations will be necessary to support
the use of results with the preliminary formulation. There Primary pharmacology
may even be situations when clinical studies must be The primary pharmacodynamic studies provide the first
repeated. evidence that the compound has the pharmacological
The analytical methods used and their validation must effects required to give therapeutic benefit. It is a clear
be described. Manufacturing processes and their valida- regulatory advantage to use established models and to be
tion are also required to demonstrate interbatch uniform- able at least to establish a theory for the mechanism of
ity. However, full-scale validation may be submitted when action. This will not always be possible and is not a firm
sales production has eventually started. requirement, but proof of efficacy and safety is helped by
Studies on the stability of both substance and products a plausible mechanistic explanation of the drug’s effects,
under real-life conditions are required, covering the full and this knowledge will also become a very powerful tool
time of intended storage. Preliminary stability data are for marketing. For example, understanding the mecha-
sufficient for the start of clinical studies. The allowable nism of action of proton pump inhibitors, such as
storage time can be increased as data are gathered and omeprazole (see Chapter 4), was important in explaining
submitted. Even marketing authorizations can be approved their long duration of action, allowing once-daily dosage
on less than real-time storage information, but there is a of compounds despite their short plasma half-life.
requirement to submit final data when available.
Inactive ingredients as well as active substances need to
be documented, unless they are well known and already General pharmacology
documented. Even then it may become necessary to General pharmacology1 studies investigate effects other
perform new animal studies to support novel uses of com- than the primary therapeutic effects. Safety pharmacology
monly used additives. studies (see Chapter 15), which must conform to good
Although the quality module of the documentation is laboratory practice (GLP) standards, are focused on iden-
the smallest, the details of requirements and the many tifying the effects on physiological functions that in a
changes needed during development and maintenance of clinical setting are unwanted or harmful.
a product make it the most resource intensive module
from a regulatory perspective. Also, legislation differs most
1
in this area, so it will often be necessary to adapt the docu- There are a number of widely used terms with similar meanings,
e.g. secondary pharmacology, safety pharmacology, high-dose
mentation for the intended regional submission. RA pro- pharmacology, regulatory pharmacology and pharmacodynamic
fessionals, however, try to convince regulatory authorities safety.

289
Section | 3 | Drug Development

Although the study design will depend on the properties Toxicology


and intended use of the compound, general pharmacol-
The principles and methodology of toxicological assess-
ogy studies are normally of short duration (i.e. acute,
ment of new compounds are described in Chapter 15.
rather than chronic, effects are investigated), and the
Here we consider the regulatory aspects.
dosage is increased until clear adverse effects occur. The
In contrast to the pharmacological studies, toxicological
studies also include comparisons with known compounds
studies generally follow standard protocols that do not
whose pharmacological properties or clinical uses are
depend on the compound characteristics. Active compara-
similar.
tors are not used, but the drug substance is compared at
When required, e.g. when pharmacodynamic effects
various dose levels to a vehicle control, given, if possible,
occur only after prolonged treatment, or when effects seen
via the intended route of administration.
with repeated administration give rise to safety concerns,
the duration of a safety pharmacology study needs to be
Single and repeated-dose studies
prolonged. The route of administration should, whenever
possible, be the route intended for clinical use. The acute toxicity of a new compound must be evaluated
There are cases when a secondary pharmacological prior to the first human exposure.
effect has, eventually, been developed into a new indica- This information is obtained from dose escalation
tion. Lidocaine, for example, was developed as a local studies or dose-ranging studies of short duration. Lethality
anaesthetic agent and its cardiac effects after overdose were is no longer an ethically accepted endpoint. The toxicol-
considered a hazard. Later that cardiac effect was exploited ogy requirements for the first exploratory studies in man
as a treatment for ventricular arrhythmia. are described in the ICH guidline M3 which reached step
All relevant safety pharmacology studies must be 5 in June 2009 (see website reference 1). Table 20.1 shows
completed before studies can be undertaken in patients. the duration of repeated-dose studies recommended by
Complementary studies may still be needed to clarify ICH, to support clinical trials and therapeutic use for dif-
unexpected findings in later development stages. ferent periods.

Genotoxicity
Pharmacokinetics: absorption, distribution, Preliminary genotoxicity evaluation of mutations and
metabolism and excretion (ADME) chromosomal damage (see Chapter 15) is needed before
Preliminary pharmacokinetic tests to assess the absorp- the drug is given to humans. If results from those studies
tion, plasma levels and half-life (i.e. exposure information) are ambiguous or positive, further testing is required. The
are performed in rodents in parallel with the preliminary entire standard battery of tests needs to be completed
pharmacology and toxicology studies (see Chapter 10). before Phase II (see Chapter 17).
Studies in humans normally start with limited short-
term data, and only if the results are acceptable are detailed Carcinogenicity
animal and human ADME studies performed. The objective of carcinogenicity studies is to identify any
Plasma concentrations observed in animals are used to tumorigenic potential in animals, and they are required
predict the concentrations that may be efficacious/ only when the expected duration of therapy, whether con-
tolerated in humans, under the assumption that similar tinuous or intermittent, is at least 6 months. Examples
biological effects should be produced at similar plasma include treatments for conditions such as allergic rhinitis,
levels across species. This is a reasonable assumption pro- anxiety or depression.
vided the in vitro target affinity is similar. Carcinogenicity studies are also required when there is
Investigations during the toxicology programme give particular reason for concern, such as chemical similarities
the bulk of the pharmacokinetic information due to the to known carcinogens, pathophysiological findings in
long duration of drug exposure and the wide range of animal toxicity studies, or positive genotoxicity results.
doses tested in several relevant species. They also give data Compounds found to be genotoxic by in vitro as well as
about tissue distribution and possible accumulation in the in vivo tests are presumed to be trans-species carcinogens
body, including placental transfer and exposure of the with hazards to humans.
fetus, as well as excretion in milk. Carcinogenicity studies normally run for the lifespan of
Metabolic pathways differ considerably between species, the test animals. They are performed quite late in the
often quantitatively but sometimes also qualitatively. development programme and are not necessarily com-
Active metabolites can influence study results, in particular pleted when the application for marketing authorization
after repeated use. A toxic metabolite with a long half-life is submitted. Indeed, for products for which there is a
may accumulate in the body and disturb results. The char- great medical need in the treatment of certain serious
acterization and evaluation of metabolites are long proc- diseases, the regulatory authority may agree that submis-
esses, and are generally the last studies to be completed in sion of carcinogenicity data can be delayed until after
a development programme. marketing approval is granted.

290
Regulatory affairs Chapter | 20 |

Table 20.1  Duration of repeated-dose toxicity studies

Recommended minimum duration of repeated-dose toxicity studies

Maximum duration To support clinical trials To support marketing


of clinical trials

Rodents Non-rodents Rodents Non-rodents


Up to 2 weeks 2 weeksa 2 weeks 1 month 1 month
>2 weeks to 1 month Same as the clinical trialb Same as the clinical trial b
3 months 3 months
>1 month to 3 months 6 months 3 months
>3 months to 6 months 6 months c
9 monthscd
>6 months 6 monthsbc 9 monthsbcd
a
In the USA, as an alternative to 2-week studies, single-dose toxicity studies with extended examination can support single-dose human
trials.
b
Data from 3-month studies in rodents and non-rodents may be sufficient to start clinical trials longer than 3 months provided longer-term
data are made available before extension of the clinical trial.
c
If paediatric patients are the target population, long-term toxicity studies in juvenile animals may be required.
d
6 months’ studies are acceptable in non-rodents in certain cases, e.g. intermittent treatment of migraine, chronic treatment to prevent
recurrence of cancer, indications for which life expectancy is short, animals cannot tolerate the treatment.

Reproductive and developmental toxicity Box 20.1  Requirement for reproduction toxicity
These studies (see Chapter 15) are intended to reveal related to clinical studies in fertile women
effects on male or female fertility, embryonic and fetal
development, and peri- and postnatal development. EU: Embryo/fetal development studies are
An evaluation of effects on the male reproductive system required before Phase I, and female fertility
is performed in the repeated-dose toxicity studies, and this should be completed before Phase III
histopathological assessment is considered more sensitive USA: Careful monitoring and pregnancy testing
in detecting toxic effects than are fertility studies. Men can may allow fertile women to take part
therefore be included in Phase I–II trials before the male before reproduction toxicity is available.
fertility studies are performed in animals. Female fertility and embryo/fetal assessment
Women may enter early studies before reproductive tox- to be completed before Phase III
icity testing is completed, provided they are permanently Japan: Embryo/fetal development studies are
sterilized or menopausal, and provided repeated-dose tox- required before Phase I, and female fertility
icity tests of adequate duration have been performed, should be completed before Phase III.
including the evaluation of female reproductive organs.
For women of childbearing potential there is concern
regarding unintentional fetal exposure, and there are
regional differences (Box 20.1) in the regulations about Efficacy assessment (studies in man)
including fertile women in clinical trials. When the preclinical testing is sufficient to start studies in
man, the RA department compiles a clinical trials submis-
Local tolerance and other toxicity studies sion, which is sent to the regulatory authority and the
The purpose of local tolerance studies is to ascertain ethics committee (see Regulatory procedures, below).
whether medicinal products (both active substances and The clinical studies, described in detail in Chapter 17,
excipients) are tolerated at sites in the body that may come are classified according to Table 20.2.
into contact with the product in clinical use. This could
mean, for example, ocular, dermal or parenteral adminis-
tration. Other studies may also be needed. These might be Human pharmacology
studies on immunotoxicity, antigenicity studies on meta­ Human pharmacology studies refer to the earliest human
bolites or impurities, and so on. The drug substance and exposure in volunteers, as well as any pharmacological
the intended use will determine the relevance of other studies in patients and volunteers throughout the develop-
studies. ment of the drug.

291
Section | 3 | Drug Development

Table 20.2  ICH classification of clinical studies

Type of Study objectives Traditional


study terminology
Human Assess tolerance; describe or define pharmacokinetics/pharmacodynamics; Phase I
pharmacology explore drug metabolism and drug interactions; estimate activity
Therapeutic Explore use for the targeted indication; estimate dosage for subsequent Phase II
exploratory studies; provide basis for confirmatory study design, endpoints, methodologies
Therapeutic Demonstrate or confirm efficacy; establish safety profile; provide an adequate Phase III
confirmatory basis for assessing benefit–risk relationship to support licensing (drug (a and b)
approval); establish dose–response relationship
Therapeutic use Refine understanding of benefit–risk relationship in general or special Phase IV
populations and/or environments; identify less common adverse reactions;
refine dosing recommendations

The first study of a new drug substance in humans has factors such as concomitant drug treatment, diet, social
essentially three objectives: habits (e.g. tobacco or alcohol), age, gender, ethnic origin
• To investigate tolerability over a range of doses and, if and time of administration.
possible, see the symptoms of adverse effects Interaction studies can be performed in healthy volun-
• To obtain information on pharmacokinetics, and to teers looking at possible metabolism changes when
measure bioavailability and plasma concentration/ co-administering compounds that share the same enzy-
effect relations matic metabolic pathway. Also, changed pharmacokinetic
• To examine the pharmacodynamic activity over a range behaviour can be investigated in combinations of drugs
of doses and obtain a dose–response relationship, that are expected to be used together. Generally, such
provided a relevant effect can be measured in healthy human volunteer studies are performed when clinical
volunteers. findings require clarification or if the company wishes to
avoid standard warning texts which would otherwise
Further human pharmacology studies are performed
apply to the drug class.
to document pharmacodynamic and pharmacokinetic
effects. Examples of the data needed to support an applica-
tion for trials in patients are the complete pharmacoki- Therapeutic exploratory studies
netic evaluation and the performance of bioavailability/ After relevant information in healthy volunteers has
bioequivalence studies during the development of new been obtained, safety conclusions from combined animal
formulations or drug-delivery systems. Information is also and human exposure will be assessed internally. If these
obtained on the possible influence of food on absorption, are favourable, initial patient studies can begin. To obtain
and that of other concomitant medications, i.e. drug inter- the most reliable results, the patient population should
action. Exploration of metabolic pattern is also performed be as homogeneous as possible – similar age, no other
early in the clinical development process. diseases than the one to be studied – and the design
Special patient populations need particular attention should, when ethically justified, be placebo controlled.
because they may be unduly sensitive or resistant to treat- For ethical reasons only a limited number of closely
ment regimens acceptable to the normal adult population monitored patients take part in these studies. Their impor-
studied. One obvious category is patients with renal or tance lies in the assumption that any placebo effect in the
hepatic impairment, who may be unable to metabolize or group treated with active drug should be eliminated by
excrete the drug effectively enough to avoid accumulation. comparison with blinded inactive treatment. They are
The metabolic pattern and elimination route are impor- used primarily to establish efficacy measured against no
tant predictors for such patients, who are not included in treatment.
clinical trials until late in development.
Gender differences may also occur, and may be detected
Studies in special populations: elderly,
by the inclusion of women at the dose-finding stage of
clinical trials. children, ethnic differences
An interaction is an alteration in the pharmacodynamic Clinically significant differences in pharmacokinetics
or the pharmacokinetic properties of a drug caused by between the elderly and the young are due to several

292
Regulatory affairs Chapter | 20 |

factors related to aging, such as impaired renal function, Ethnic differences


which can increase the variability in drug response, as well
In order for clinical data to be accepted globally, the risk
as increasing the likelihood of unwanted effects and drug
of ethnic differences must be assessed. ICH Efficacy guide-
interactions. Bearing in mind that the elderly are the
line E5 defines a bridging data package that would allow
largest group of consumers, this category should be studied
extrapolation of foreign clinical data to the population in
as early as possible in clinical trials.
the new region. A limited programme may suffice to
Clinical trials in children confirm comparable effects in different ethnic groups.
Ethnic differences may be genetic in origin, as in the
Studies in children require experience from adult human
example described in Chapter 17, or related to differences
studies and information on the pharmacokinetic profile
in environment, culture or medical practice.
of the substance. Because of the difficulties, and the often
small commercial return, companies have seldom consid-
ered it worthwhile to test drugs in children and to seek Therapeutic confirmatory studies
regulatory approval for marketing drugs for use in chil- The therapeutic confirmatory phase is intended to confirm
dren. Nevertheless, drugs are often prescribed ‘off-label’ efficacy results from controlled exploratory efficacy studies,
for children, on the basis of clinical experience suggesting but now in a more realistic clinical environment and with
that they are safe and effective. Such off-label prescribing a broader population.
is undesirable, as clinical experience is less satisfactory In order to convincingly document that the product is
than formal trials data as a guide to efficacy and safety, efficacious, there are some ‘golden rules’ to be aware of in
and because it leaves the clinician, rather than the phar- terms of the need for statistical power, replication of the
maceutical company, liable for any harm that may result. results, etc. Further, for a product intended for long-term
Recently, requirements to include a paediatric population use, its performance must usually be investigated during
early have forced the development of new guidance as to long-term exposure. All these aspects will, however, be
how to include children in clinical development. Market influenced by what alternative treatments there are, the
exclusivity prolongation has been successfully tried for intended target patient population, the rarity and severity
some years in the USA, and in July 2003 the Federal Food of the disease as well as other factors, and must be evalu-
Drug and Cosmetics Act was amended to request paediat- ated case by case.
ric studies in a new submission unless omission is justi-
fied. In September of 2007, the Paediatric Research Equity
Act replaced the 2003 Act and an assessment was made to
Clinical safety profile
evaluate the quality of the studies submitted. The results An equally important function of this largest and longest
were positive and between September 2007 and June 2010 section of the clinical documentation is to capture all
more than 250 clinical trials comprising more than adverse event information to enable evaluation of the rela-
100 000 children have been performed. (see website refer- tive benefit–risk ratio of the new compound, and also to
ence 2). detect rare adverse reactions. To document clinical safety,
In Europe the European Commission adopted the Pae- the ICH E1 guideline on products intended for chronic use
diatric Regulation in February 2007 (see website reference stipulates that a minimum of 100 patients be treated for
3). This stipulates that no marketing approval will be at least 1 year, and 300–600 treated for at least 6 months.
granted unless there is an agreed paediatric investigation In reality, however, several thousand patients usually form
plan in place or, alternatively, there is a waiver from the the database for safety evaluation for marketing approval.
requirement because of the low risk that the product will Not until several similar studies can be analysed together
be considered for use in children. The paediatric studies can a real estimate be made of the clinical safety of the
may, with the agreement of the regulatory authority, be product.
performed after approval for other populations. For new The collected clinical database should be analysed
compounds or for products with a Supplementary Protec- across a sensible selection of variables, such as sex, age,
tion Certificate (see p. 300), paediatric applications will race, exposure (dose and duration), as well as concomitant
be given a longer market exclusivity period, and off-patent diseases and concomitant pharmacotherapy This type of
products will be given a special paediatric use marketing integrated data analysis is a rational and scientific way to
authorization (PUMA); funds will be available for paedi- obtain necessary information about the benefits and risks
atric research in these products. of new compounds, and has been required for FDA sub-
To further encourage paediatric research, authority sci- missions for many years, though it is not yet a firm require-
entific advice is free. Paediatric clinical data from the EU ment in the EU or Japan.
and elsewhere are collected in a common European data- To further emphasize the accountability for the product
base to avoid repetition of trials with unnecessary expo- by the pharmaceutical company a more proactive legisla-
sure in children. The US FDA and the EMA in Europe tion for risk evaluation has been introduced in USA and
exchange information on paediatric clinical research. EU. The term Risk Evaluation and Mitigation Strategy

293
Section | 3 | Drug Development

(REMS) in the USA is matched by Risk Management stringent, but procedures and standards are flexible and
System in EU. generally established on a case-by-case basis. Conse-
The difference from the established method of reporting quently, achieving regulatory approval can often be a
adverse reactions that have occurred is that any possible greater challenge for the pharmaceutical company, but
or potential risks for a patient being harmed should be there are also opportunities to succeed with novel and
foreseen or identified early and mitigation/minimization relatively quick development programmes.
activities be planned in advance. The risk management Published guidelines on the development of conven-
document is generally part of the regulatory approval and tional drugs need to be considered to determine what parts
follows the product’s entire lifecycle. The true risk profile are relevant for a particular biopharmaceutical product. In
will develop along with the increased knowledge base. The addition, there are, to date, seven ICH guidelines dealing
goal is at any time to be able to demonstrate how and why exclusively with biopharmaceuticals, as well as numerous
the treatment benefits outweigh the risks (see website ref- FDA and CHMP guidance documents. These mostly deal
erences 4 and 5). with quality aspects and, in some cases, preclinical safety
aspects. The definition of what is included in the term
‘biopharmaceutical’ varies somewhat between documents,
Regulatory aspects of novel types
and therefore needs to be checked. The active substances
of therapy include proteins and peptides, their derivatives, and prod-
As emphasized in earlier chapters, the therapeutic scene is ucts of which they are components. Examples include (but
moving increasingly towards biological and biopharma- are not limited to) cytokines, recombinant plasma factors,
ceutical treatments, as well as various innovative, so-called growth factors, fusion proteins, enzymes, hormones and
advanced therapies. There are also broadening definitions monoclonal antibodies (see also Chapters 12 and 13).
of what constitutes medical devices. The regulatory frame-
Quality considerations
work established to ensure the quality, safety and efficacy
of conventional synthetic drugs is not entirely appropriate A unique and critically important feature for biopharma-
for many biopharmaceutical products, and even less so for ceuticals is the need to ensure and document viral safety
the many gene- and cell-based products currently in devel- aspects. Furthermore, there must be preparedness for
opment. Recombinant proteins have been in use since potentially new hazards, such as infective prions. There-
1982 and the regulatory process for such biopharmaceu- fore strict control of the origin of starting materials and
ticals is by now well established, hence this chapter will expression systems is essential. The current battery of ICH
mainly focus on these. The EU also has, for example, new quality guidance documents in this area reflects these
legislation in force since December 2008 specifically on points of particular attention (ICH Q5A-E, and Q6B).
Advanced Therapy Medicinal Products (ATMP), meaning At the time when biopharmaceuticals first appeared, the
somatic cell therapy, gene therapy and tissue engineering. ability to analyse and exactly characterize the end-product
This involves requirements for the applicant, as part of a was not possible the way it was with small molecules.
marketing authorization, to establish a risk management Therefore, their efficacy and safety depended critically on
system. In addition to the standard requirements in terms the manufacturing process itself, and emphasis was placed
of safety follow-up for an approved product, this system on ‘process control’ rather than ‘product control’.
should also include an evaluation of the product’s effec- Since then, much experience and confidence has been
tiveness (see website reference 6). In the USA, there are a gained. Bioanalytical technologies for characterizing large
number of FDA guidelines relating to cellular and gene molecules have improved dramatically and so has the field
therapies. The regulatory framework for even newer thera- of bioassays, which are normally required to be included
peutic modalities relating, for example, to nanotechnolo- in such characterizations, e.g. to determine the ‘potency’
gies is not yet clearly defined, and the regulatory authorities of a product. As a result, the quality aspects of biophar-
face a difficult task in keeping up with the rapid pace of maceuticals are no longer as fundamentally different from
technological change. those of synthetic products as they used to be. The quality
documentation will still typically be more extensive than
it is for a small-molecule product.
Biopharmaceuticals Today the concept of ‘comparability’ has been estab-
Compared with synthetic compounds, biopharmaceuti- lished and approaches for demonstrating product compa-
cals are by their nature more heterogeneous, and their rability after process changes have been outlined by
production methods are very diverse, including complex regulatory authorities. Further, the increased understand-
fermentation and recombinant techniques, as well as pro- ing of these products has in more recent times allowed the
duction via the use of transgenic animals and plants, approval of generic versions also of biopharmaceuticals,
thereby posing new challenges for quality control. This has so-called follow-on biologics or biosimilars. A legal frame-
necessarily led to a fairly pragmatic regulatory framework. work was established first in the EU, in 2004, and the first
Quality, safety and efficacy requirements have to be no less such approvals (somatropin products) were seen in 2006.

294
Regulatory affairs Chapter | 20 |

Approvals are so far slightly behind in the USA since there Efficacy considerations
was actually not a regulatory pathway for biosimilars until
The need to establish efficacy is in principle the same for
such provisions were signed into law in March 2010 (see
biopharmaceuticals as for conventional drugs, but there
website reference 7). The FDA is, at the time of writing,
are significant differences in practice. The establishment of
working on details of how to implement these provisions
a dose–response relationship can be irrelevant, as there
to approve biosimilars. Also, in Japan, the authorities have
may be an ‘all-or-none-effect’ at extremely low levels. Also,
published ‘Guidelines for the Quality, Safety and Efficacy
to determine a maximum tolerated dose (MTD) in humans
Assurance of Follow-on Biologics’, the most recent version
may be impractical, as many biopharmaceuticals will not
being in March 2009.
evoke any dose-limiting side effects. Measuring pharma-
Safety considerations cokinetic properties may be difficult, particularly if the
substance is an endogenous mediator. Biopharmaceuticals
The expectations in terms of performing and documenting may also have very long half-lives compared to small mol-
a non-clinical safety evaluation for biotechnology-derived ecules, often in the range of weeks rather than hours.
pharmaceuticals are well outlined in the ICH guidance For any biopharmaceutical intended for chronic or
document S6. This guideline was revised in 2011 driven by repeated use, there will be extra emphasis on demonstrat-
scientific advances and experience gained since publica- ing long-term efficacy. This is because the medical use of
tion of the original guidance. It indicates a flexible, case- proteins is associated with potential immunogenicity and
by-case and science-based approach, but also points out the possible development of neutralizing antibodies, such
that a product needs to be sufficiently characterized to that the intended effect may decrease or even disappear
allow the appropriate design of a preclinical safety with time. Repeated assessment of immunogenicity may
evaluation. be needed, particularly after any process changes.
Generally, all toxicity studies must be performed accord- To date, biopharmaceuticals have typically been devel-
ing to GLP. However, for biopharmaceuticals it is recog- oped for serious diseases, and certain types of treatments,
nized that some specialized tests may not be able to such as cytotoxic agents or immunomodulators, cannot be
comply fully with GLP. The guidance further comments given to healthy volunteers. In such cases, the initial dose-
that the standard toxicity testing designs in the commonly escalation studies will have to be carried out in a patient
used species (e.g. rats and dogs) are often not relevant. population rather than in normal volunteers.
To make it relevant, a safety evaluation should include
a species in which the test material is pharmacologically Regulatory procedural considerations
active. Further, in certain justified cases one relevant
In the EU, only the centralized procedure can be used for
species may suffice, at least for the long-term studies. If no
biopharmaceuticals as well as biosimilars (see Regulatory
relevant species at all can be identified, the use of trans-
procedures, below). In the USA, biopharmaceuticals are in
genic animals expressing the human receptor or the use of
most cases approved by review of Biologics License Appli-
homologous proteins should be considered.
cations (BLA), rather than New Drug Applications (NDA).
Other factors of particular relevance with biopharmaceu-
ticals are potential immunogenicity and immunotoxicity.
Long-term studies may be difficult to perform, depending
Personalized therapies
on the possible formation of neutralizing antibodies in the It is recognized that an individual’s genetic makeup influ-
selected species. For products intended for chronic use, the ences the effect of drugs. Incorporating this principle into
duration of long-term toxicity studies must, however, therapeutic practice is still at a relatively early stage,
always be scientifically justified. Regulatory guidance also although the concept moves rapidly towards everyday
states that standard carcinogenicity studies are generally reality, and it presents a challenge for regulatory authori-
inappropriate, but that product-specific assessments of ties who are seeking ways to incorporate personalized
potential risks may still be needed, and that a variety of medicine (pharmacogenomics) into the regulatory
approaches may be necessary to accomplish this. process. How a drug product can or should be co-developed
In 2006 disastrous clinical trial events took place with with the necessary genomic biomarkers and/or assays is
an antibody (TGN 1412), where healthy volunteers suf- not yet clear. Another regulatory uncertainty has been how
fered life-threatening adverse effects. These effects had not the generation of these kinds of data will translate into
been anticipated by the company or the authority review- label language for the approved product. Already in 2005
ing the clinical trial application. The events triggered the FDA issued guidance on a specific procedure for ‘Phar-
intense discussions on risk identification and mitigation macogenomic Data Submissions’ to try and alleviate
for so-called first-in-human clinical trials, and in particular potential industry concerns and instead promote scientific
for any medicinal product which might be considered progress and the gaining of experience in the field. In the
‘high-risk’. Since then, specific guidelines for such clinical EU there is, at the time of writing, draft guidance pub-
trial applications have been issued by the regulatory lished on the use of pharmacogenetic methodologies in
authorities. the pharmacokinetic evaluation of medicinal products.

295
Section | 3 | Drug Development

However, still today, there are relatively few approved however, be excluded from this requirement under certain
products to learn from. conditions (see website reference 10, p. 301), for example
if the estimated concentration in the aquatic environment
of the active substance is below 1 part per billion, or if the
Orphan drugs
substance occurs naturally. In Europe a corresponding
Orphan medicines are those intended to diagnose, prevent general guideline was implemented in December 2006 for
or treat rare diseases, in the EU further specified as life- human medicinal products (see website reference 11).
threatening or chronically debilitating conditions. The
concept also includes therapies that are unlikely to be
developed under normal market conditions, where the REGULATORY PROCEDURES
company can show that a return on research investment
will not be possible. To qualify for orphan drug status,
there should be no satisfactory treatment available or, alter- Clinical trials
natively, the intended new treatment should be assumed It is evident that clinical trials can pose unknown risks to
to be of significant benefit (see website references 8 and 9) humans: the earlier in the development process, the
To qualify as an orphan indication, the prevalence of greater the risk. Regulatory and ethical approvals are based
the condition must be fewer than 5 in 10 000 individuals on independent evaluations, and both are required before
in the EU, fewer than 50 000 affected in Japan, or fewer investigations in humans can begin.
than 200 000 affected in the USA. Financial and scientific The ethical basis for all clinical research is the Declara-
assistance is made available for products intended for use tion of Helsinki (see website reference 12, p. 301), which
in a given indication that obtain orphan status. Examples states that the primary obligation for the treating physician
of financial benefits are a reduction in or exemption from is to care for the patient. It also says that clinical research
fees, as well as funding provided by regulatory authorities may be performed provided the goal is to improve treat-
to meet part of the development costs in some instances. ment. Furthermore, the subject must be informed about
Specialist groups within the regulatory authorities provide the potential benefits and risks, and must consent to par-
scientific help and advice on the execution of studies. ticipate. The guardians of patients who for any reason
Compromises may be needed owing to the scarcity of cannot give informed consent (e.g. small children) can
patients, although the normal requirements to demon- agree on participation.
strate safety and efficacy still apply. Regulatory authorities are concerned mainly with the
The most important benefit stimulating orphan drug scientific basis of the intended study protocol and of
development is, however, market exclusivity for 7–10 years course the safety of subjects/patients involved. Regulatory
for the product, for the designated medical use. In the EU authorities require all results of clinical trials approved by
the centralized procedure (see Regulatory procedures, them to be reported back.
below) is the compulsory procedure for orphan drugs. All clinical research in humans should be performed
The orphan drug incentives are fairly recent. In the USA according to the internationally agreed Code of Good
the legislation dates from 1983, in Japan from 1995, and Clinical Practice, as described in the ICH guideline E6 (see
in the EU from 2000. The US experience has shown very website reference 1, p. 301).
good results; a review of the first 25 years of the Orphan
Drug Act resulted in 326 marketing approvals, the majority
of which are intended to treat rare cancers or metabolic/ Europe
endocrinological disorders. Until recently, the regulatory requirements to start and
conduct clinical studies in Europe have varied widely
between countries, ranging from little or no regulation to
Environmental considerations
a requirement for complete assessment by the health
Environmental evaluation of the finished pharmaceutical authority of the intended study protocol and all support-
products is required in the USA and EU, the main concern ing documentation.
being contamination of the environment by the com- The efforts to harmonize EU procedures led to the devel-
pound or its metabolites. The environmental impact of the opment of a Clinical Trial Directive implemented in May
manufacturing process is a separate issue that is regulated 2004 (see website reference 13, p. 301). One benefit is
elsewhere. that both regulatory authorities and ethics committees
The US requirement for environmental assessment must respond within 60 days of receiving clinical trials
(EA) applies in all cases where action is needed to mini- applications and the requirements for information to be
mize environmental effects. An Environmental Assess- submitted are being defined and published in a set of
ment Report is then required, and the FDA will develop guidelines. Much of the information submitted to the
an Environmental Impact Statement to direct necessary regulatory authority and ethics committee is the same. In
action. Drug products for human or animal use can, spite of the Clinical Trial Directive, however, some national

296
Regulatory affairs Chapter | 20 |

differences in format and information requirements have The requirements for beginning a clinical study in Japan
remained, and upon submission of identical clinical are similar to those in the USA or Europe. Scientific
trial protocols in several countries the national reviews summary information is generally acceptable, and ethics
may reach different conclusions. Therefore, at the time of committee approval is necessary.
writing, there is a pilot Voluntary Harmonization Proce-
dure (VHP) available for applications involving at least
three EU member states. This comprises two steps, where Application for marketing
in the first round one coordinator on the regulatory authorization
authority side manages national comments on the appli-
cation and provides the applicant with a consolidated list The application for marketing authorization (MAA in
of comments or questions. In the second step the usual Europe, NDA in the USA, JNDA in Japan) is compiled and
national applications for the clinical trial are submitted submitted as soon as the drug development programme
but national approval should then be gained very rapidly has been completed and judged satisfactory by the
without any further requests for change. company. Different authorities have differing require-
ments as to the level of detail and the format of submis-
sions, and it is the task of the RA department to collate all
USA the data as efficiently as possible to satisfy these varying
An Investigational New Drug (IND) application must be requirements with a minimum of redrafting.
submitted to the FDA before a new drug is given to The US FDA in general requires raw data to be submit-
humans. The application is relatively simple (see website ted, allowing them to make their own analysis, and thus
reference 14, p. 301), and to encourage early human they request the most complete data of all authorities.
studies it is no longer necessary to submit complete phar- European authorities require a condensed dossier contain-
macology study reports. The toxicology safety evaluation ing critical evaluations of the data, allowing a rapid review
can be based on draft and unaudited reports, provided the based on conclusions drawn by named scientific experts.
data are sufficient for the FDA to make a reliable assess- These may be internal or external, and are selected by the
ment. Complete study reports must be available in later applicant.
clinical development phases. Japanese authorities have traditionally focused predom-
Unless the FDA has questions, or even places the product inantly on data generated in Japan; studies performed
on ‘clinical hold’, the IND is considered opened 30 days elsewhere being supportive only.
after it was submitted and from then on information Below, the procedures adopted in these three regions are
(study protocol) is added to it for every new study planned. described in more detail.
New scientific information must be submitted whenever
changes are made, e.g. dose increases, new patient catego- Europe
ries or new indications. The IND process requires an
annual update report describing project progress and addi- Several procedures are available for marketing authoriza-
tional data obtained during the year. tion in the EU (Figure 20.3, see website reference 13):
Approval from Institutional Review Board (IRB) is • National procedure, in which the application is
needed for every institution where a clinical study is to be evaluated by one regulatory authority. This procedure
performed. is allowed for products intended for that country
only. Also, it is the first step in a mutual recognition
procedure.
Japan
• Mutual recognition, in which a marketing approval
Traditionally, because of differences in medical culture, in application is assessed by one national authority, the
treatment traditions and the possibility of significant racial Reference Member State (RMS), which subsequently
differences, clinical trials in Japan have not been useful for defends the approval and evaluation in order to
drug applications in the Western world. Also, for a product gain mutual recognition of the assessment from
to be approved for the Japanese market, repetition of clini- other European authorities. The pharmaceutical
cal studies in Japan was necessary, often delaying the avail- company may select countries of interest. These,
ability of products in Japan. the Concerned Member States, have 90 days to
With the introduction of international standards under recognize the initial assessment. The Mutual
the auspices of ICH, data from Japanese patients are Recognition procedure is used for harmonization
increasingly becoming acceptable in other countries. The and conversion of nationally approved products.
guideline on bridging studies to compensate for ethnic After mutual recognition the final marketing
differences (ICH E5; see website reference 1, p. 301) authorizations are given as national decisions, but
may allow Japanese studies to become part of global the scientific assessment is quicker and requires
development. fewer resources from all national authorities. In the

297
Section | 3 | Drug Development

After close of procedure, national approval in each CMS within 30 days

Submission in
Applicant
RMS and CMS

For info

Day 70, To arbitration


Preliminary
Assessment
report
No concensus
For response

Assessment step II Day 210


Day 100, Close of procedure
Consultation if consensus
RMS, CMS, No
Applicant Consensus

Clock stop, 3 months Until Day 120


RMS prepares Day 120,
Close of procedure
Draft Assessment Consensus To National approval
Report

RMS = Reference Member State, CMS = Concerned Member State

Fig. 20.3  One of the submission processes for marketing approval in EU, the Decentralized Procedure.

case of non-agreement, referral to EMA for for any reason is not submitted to follow the
arbitration is done as a last resort. But before centralized procedure.
arbitration is considered, the Coordination Group • Centralized procedure is a ‘federal’ procedure carried
for Mutual Recognition and Decentralised Procedures out by the EMA, with scientists selected from CHMP
(CMDh/CMDv) will try to resolve outstanding to perform the review, the approval body being the
issues. This is a group composed of expert members European Commission. This procedure is mandatory
from European regulatory authorities. The worst for biotechnological products, biosimilars, orphan
outcome of an arbitration would be that the drugs as well as products intended to treat diabetes,
marketing authorizations obtained are withdrawn in AIDS, cancer, neurodegenerative disorders,
all EU countries, including the RMS. autoimmune diseases and viral diseases.
• Decentralized procedure is similar to the Mutual • The centralized procedure starts with the nomination
Recognition Procedure. It is a modernization the aim of one CHMP member to act as rapporteur, who
of which is to share the work among authorities selects and leads the assessment team. A selected
earlier in the process, with the possibility of a co-rapporteur and team make a parallel review. The
decision being reached before 120 days have European Commission approves the application
passed from receipt of a valid submission. It is the based on a CHMP recommendation, which in turn
procedure of choice for a new chemical entity that is based on the assessment reports by the two

298
Regulatory affairs Chapter | 20 |

rapporteur teams. Products approved in this way can is made by the MHLW based on the Evaluation Centre’s
be marketed in all EU countries with the same report.
prescribing information, packs and labels. It is worth mentioning that health authorities, in par-
CHMP is prepared to give scientific advice to companies ticular in the ICH regions, have well-established commu-
in situations where published guidance on the European nications and often assist and consult each other.
position is not available, or when the company needs to
discuss a possible deviation from guidelines. Such advice, The common technical document
as well as advice from national regulatory authorities, may
be very valuable at any stage of the development pro- Following the good progress made by ICH in creating
gramme, and may later be incorporated in new guidelines. scientific guidelines applicable in the three large regions,
Providing advice requires considerable effort from CHMP discussions on standardizing document formats began in
specialists, and fees have to be paid by the pharmaceutical 1997. The aim was to define a standard format, called the
company. Common Technical Document (CTD), for the application
for a new drug product. It was realized from the outset
that harmonization of content could not be achieved,
USA owing to the fundamental differences in data require-
The FDA is more willing than other large authorities to ments and work processes between different regulatory
take an active part in planning the drug development authorities. Adopting a common format would, nonethe-
process. Some meetings between the FDA and the spon- less, be a worthwhile step forward.
soring company are more or less compulsory. One such The guideline was adopted by the three ICH regions in
example is the so-called end-of-Phase II meeting. This is November 2000 and subsequently implemented, and it
in most cases a critically important meeting for the has generally been accepted in most other countries. This
company in which clinical data up until that point is saves much time and effort in reformatting documents for
presented to the FDA together with a proposed remaining submission to different regulatory authorities. The struc-
phase III programme and the company’s target label. The ture of the CTD (see website reference 1, p. 301) is sum-
purpose is to gain FDA feedback on the appropriateness marized in Figure 20.4.
of the intended NDA package and whether, in particular, Module 1 (not part of the CTD) contains regional infor-
the clinical data are likely to enable approval of a desirable mation such as the application form, the suggested pre-
product label. It is important that the advice from the FDA scribing information, the application fee, and also other
is followed. At the same time, these discussions may make information that is not considered relevant in all territo-
it possible in special cases to deviate from guidelines by ries, such as environmental assessment (required in the
prior agreement with the FDA. Furthermore, these discus- USA and Europe, but not in Japan). Certificates of different
sion meetings ensure that the authority is already familiar regional needs are also to be found in Module 1, as well
with the project when the dossier is submitted. as patent information not yet requested in the EU.
The review time for the FDA has decreased substantially Module 2 comprises a very brief general introduction,
in the last few years (see Chapter 22). Standard reviews followed by summary information relating to quality,
should be completed within 10 months, and priority safety (i.e. non-clinical studies) and efficacy (i.e. clinical
reviews of those products with a strong medical need studies). Quality issues (purity, manufacturing process,
within 6 months. stability, etc.) are summarized in a single document of a
The assessment result is usually communicated either as maximum 40 pages. The non-clinical and clinical sum-
an approval or as a complete response letter. The latter is maries each consist of a separate overview (maximum 30
in essence a request for additional information or data pages) and summaries of individual studies. The overviews
before approval. in each area are similar to the previous EU Expert Reports
in that they present critical evaluations of the programme
performed. Detailed guidelines (see website references 1,
Japan 13 and 14), based on existing US, European and Japanese
Also in Japan the regulatory authority is today available requirements, are available to indicate what tabulated
for consultation, allowing scientific discussion and feed- information needs to be included in these summaries, and
back during the development phase. These meetings how the written summaries should be drafted. The non-
tend to follow a similar pattern to those in the USA and clinical section has been fairly non-controversial. The
EU and have made it much easier to address potential guidance is very similar to the previous US summary, with
problems well before submission for marketing approval clear instructions on how to sort the studies regarding
is made. animals, doses, durations of treatment and routes of
These changes have meant shorter review times and a administration.
more transparent process. The review in Japan is per- The clinical summary is similar to what was required
formed by an Evaluation Centre, and the ultimate decision by the FDA, incorporating many features taken from the

299
Section | 3 | Drug Development

ADMINISTRATIVE RULES

Not part Patent protection and  


of the
CTD data exclusivity
Module 1 Supplementary protection certificate
Regional
administrative During the 1980s, the time taken to develop and obtain
information 1.0 marketing approval for a new drug increased so much that
the period of market exclusivity established by the original
patent could be too short to allow the company to recoup
CTD table of contents
its R&D costs. To overcome this problem (see Chapter 19),
2.1
the EU Council in 1992 introduced rules allowing com-
CTD introduction 2.2 panies to apply for a Supplementary Protection Certificate
(SPC), matching similar legislation in the USA and Japan.
Module 2 This can prolong market protection by a maximum of
Non-clinical Clinical
5 years to give an overall period of exclusivity of up to
overview overview
Quality 2.4 2.5 15 years.
CTD
overall The application for an SPC has to be submitted within
summary Non-clinical Clinical 6 months of first approval anywhere in Europe, within or
2.3 summaries summaries outside the EU, and from then on the clock starts. It is thus
2.6 2.7 strategically important to obtain first approval in a finan-
cially important market for best revenue.
Module 3 Module 4 Module 5
Quality Non-clinical Clinical
3.0 study reports study reports Data exclusivity
4.0 5.0
Data exclusivity should not be confused with patent pro-
tection. It is a means to protect the originators data,
Fig. 20.4  Diagrammatic representation of the organization meaning that generic products cannot be approved by
of the Common Technical Document (CTD). referring to the original product documentation until the
exclusivity period has ended. For a new chemical entity
the protection may be up to 10 years, depending on region.
The generation of paediatric data may extend it even
Integrated Summaries of Efficacy (ISE) and Safety (ISS). further, between 6 and 12 months.
ISE will generally fit in the clinical summary document.
The ISS document has proved very useful in drawing con-
clusions from the clinical studies by sensible pooling and Pricing of pharmaceutical  
integration, but is too large (often more than 400 pages
products – ‘the fourth hurdle’
in itself) to be accepted in the EU and Japan. This problem
may be resolved by including the ISS as a separate report The ‘fourth hurdle’ or ‘market access’ are expressions for
in Module 5. the ever growing demand for cost-effectiveness in phar­
Modules 3–5 comprise the individual study reports. maceutical prescribing. Payers, whether health insurance
Most reports are eligible for use in all three regions, pos- companies or publicly funded institutions, now require
sibly with the exception at present of Module 3, Quality, more stringent justification for the normally high price of
which may need regional content. a new pharmaceutical product. This means that, if not in
There is a gradual shift to fully electronic submissions the original application, studies have to demonstrate that
– known as e-CTD – in place of the large quantities of the clinical benefit of a new compound is commensurate
paper which have comprised an application for marketing with the suggested price, compared to the previous therapy
approval. As this gets fully implemented experimental of choice in the country of application.
data will be lodged mainly in databases, allowing the To address this concern, studies in a clinical trials pro-
information to be exchanged between pharmaceutical gramme from Phase II onwards now normally include
companies and regulatory authorities much more easily. health economic measures (see Chapter 18). In the USA,
Guidelines on the structure and interface between data- certainly, it is advantageous to have a statement of health
bases in the industry setting and those at the authorities economic benefit approved in the package insert, to justify
are available. reimbursement. Reference pricing systems are also being

300
Regulatory affairs Chapter | 20 |

implemented in Europe. A favourable benefit–risk evalua- EU European Union


tion is no longer sufficient: value for money must also be FDA Food and Drug Administration; the US
demonstrated in order to have a commercially successful Regulatory Authority
product (see also Chapter 21). GCP Good clinical practice
GLP Good laboratory practice
GMP Good manufacturing practice
ICH International Conference on Harmonization
LIST OF ABBREVIATIONS IND Investigational New Drug application (US)
IRB Institutional Review Board (US), equivalent
CHMP Committee for Medicinal Products for to Ethics Committee in Europe
Human Use, the new name to replace ISS Integrated Summary of Safety (US)
CPMP early 2004 JNDA Japanese New Drug Application
CTD Common Technical Document MAA Marketing Authorization Application (EU),
CMDh/ equivalent to NDA in USA
CMDv Coordination Group for Human/Veterinary MHLW Ministry of Health, Labor and Welfare (Jpn)
Medicinal Products for Mutual Recognition MTD Maximum tolerated dose
and Decentralised Procedure (EU) NDA New Drug Application (USA)
EC Ethics Committee (EU), known as NTA Notice to Applicants; EU set of
Institutional Review Board (IRB) in USA pharmaceutical regulations, directives and
eCTD Electronic Common Technical Document guidelines
EMA European Medicines Agency SPC Supplementary Protection Certificate
ERA Environmental Risk Assessment (EU) WHO World Health Organization

REFERENCES

Cartwright AC, Matthews BR. product registration. London: Taylor Meade TW. Subacute myelo-optic
Pharmaceutical product licensing and Francis; 1994. neuropathy and clioquinol. An
requirements for Europe. London: Drews J. In quest of tomorrow’s epidemiological case-history for
Ellis Horwood; 1991. medicines. New York: Springer- diagnosis. British Journal of
Cartwright AC, Matthews BR, editors. Verlag; 1999. Preventive and Social Medicine
International pharmaceutical 1975;29:157–69.

WEBSITE REFERENCES

1. http://www.ich.org/ 6. http://www.ema.europa.eu/docs/ 10. http://www.fda.gov/downloads/


2. http://www.fda.gov/Drugs/ en_GB/document_library/ Drugs/GuidanceCompliance
DevelopmentApprovalProcess/ Regulatory_and_procedural_ RegulatoryInformation/Guidances/
DevelopmentResources/ guideline/2009/10/ ucm070561
ucm049867.htm WC500006326.pdf 11. http://www.ema.europa.eu/docs/
3. http://ec.europa.eu/health/files/ 7. http://www.fda.gov/Drugs/ en_GB/document_library/
eudralex/vol-1/reg_2006_1902/ GuidanceComplianceRegulatory Scientific_guideline/2009/10/
reg_2006_1902_en.pdf Information/ucm215031.htm WC500003978.pdf
4. http://www.fda.gov/downloads/ 8. http://www.ema.europa.eu/ema/ 12. http://www.wma.net/
Drugs/GuidanceCompliance index.jsp?curl=pages/special_topics/ en/30publications/10policies/b3/
RegulatoryInformation/Guidances/ general/general_content_000034. index.html
UCM184128.pdf jsp&murl=menus/special_topics/ 13. http://ec.europa.eu/health/
5. http://www.ema.europa.eu/docs/ special_topics.jsp&mid=WC0b01ac documents//eudralex/index_en.htm
en_GB/document_library/ 058002d4eb 14. http:/www.fda.gov
Regulatory_and_procedural_ 9. http://www.fda.gov/ForIndustry/
guideline/2009/10/ DevelopingProductsforRare
WC500004888.pdf DiseasesConditions/default.htm

301

You might also like