Global Pharmaceutical and Biologics Regulatory Strategy Second Edition Sample Chapter 1

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

15

Buy the Book >>

1
Global Regulatory Strategy 2
3

By Chris Walker and Tina Soulis 4


5
Introduction erations that influence global strategy are the
organization’s financial situation or the product’s 6
The ultimate goal of a regulatory strategy is to intellectual property status. External examples
enable patient access, and it is within this context
that a regulatory professional must provide a
include the organization’s business partner or 7
investor community viewpoints.
regulatory strategy. A regulatory strategy can Global regulatory organizations must
be defined as a science-driven assessment of a provide comprehensive development plan input 8
product’s development options, key consider- throughout the product’s lifecycle. This input
ations and likely regulatory outcome. It should
span the earliest development stages through
starts in the preclinical development phase and
continues through postmarketing. Functional
9
further modifications planned postauthorization. regulatory units provide input on the likely
It should encompass key milestones and decision
points; consider regulatory objectives, hurdles,
acceptability of the planned evidence package 10
that will be generated for the regulator and
the regulatory landscape and precedents; and subsequent stakeholders, specific regulatory
characterize risks to potential success in deliver- requirements for product development, post- 11
ing a specific regulatory outcome. This regulatory marketing, regulatory submission management,
outcome, in turn, will have broader consideration
because it will link to the potential for patient
regulatory intelligence, product labeling and
advertising and promotion.
12
access, commercial acceptability and uptake and,
Interdisciplinary Factors and Alignment
therefore, likely business outcomes.
A global regulatory strategy should combine
13
regulatory requirements and business objectives. A global regulatory strategy cannot be planned
It often is defined by a global regulatory expert, in isolation from other factors contributing
to an innovative new product’s overall suc-
14
who must consult with a cross-functional team.
The cross-functional team should comprise cess. The most successful new products use a
experts who: provide regional regulatory require- multi-pronged development program approach, 15
ments and regulatory intelligence on expecta- addressing business, scientific and regulatory out-
comes. The integrated multidisciplinary product
tions, precedents and competition; correspond
with local regulatory authorities; enable docu- development concept is shown in Figure 15-1. 16
ment management and submission processes; Business Strategy
and provide specific functional expertise such as 17
labeling, CMC, nonclinical and clinical. There Business outcomes are defined broadly as those
also are internal and external business consider- accomplishments driving business results, includ-
ations that can drive a specific global regulatory ing revenue, earnings, cash flow, return on capital 18
strategy’s development. Examples of consid- and valuation.

187
Chapter 15: Global Regulatory Strategy

Figure 15-1. Multidisciplinary Product Development

Regulatory

Business Marketing
Finance

Target Product Profile


and Integrated Product
Strategy

Clinical Medical
Manufacturing Safety

Health
Outcomes

Regulatory decisions can affect the time- not be positive, and product divestiture may be
to-market, label claims and reimbursement considered. Ultimately, companies should seek
directly and, consequently, a new product’s sales, improved patient outcomes compared to existing
profitability and contribution to business results. therapies and, if these are positive, it is likely the
Therefore, it is critical regulatory professionals product also may provide financial return.
understand the organization’s business strategy Note, a sponsor TPP should be differen-
to plan the product development and postau- tiated from a formal regulatory TPP prepared
thorization activities within this framework and in accordance with FDA guidance1 and serving
as members of a multidisciplinary global team as a format for sponsor discussions with the
planning an integrated product strategy. regulatory authority. The relationship between
Early sponsor development of a Target the two types of product profiles is important,
Product Profile (sponsor TPP) by the multidis- as a sponsor TPP will guide the formal TPP
ciplinary team establishes key product attributes and development plan, contributing to overall
and provides an opportunity to consider financial business objectives.
factors and build a sophisticated financial model A sponsor TPP may include optimal and
to develop anticipated business outcomes. It acceptable label claims compared to currently
also will assist in determining the break-even available products and competitors in develop-
point at which no further development should ment. Similarly, it may articulate an acceptable
be undertaken, as the business outcomes will safety profile and risk-benefit analysis, as well as

188
Global Pharmaceutical and Biologics Regulatory Strategy, Second Edition

1
planned product presentation. The sponsor TPP positive, but more data are required to confirm
will establish the global development plan frame- benefit post-license. 2
work, incorporating data and regulatory require- Conversely, local regulations requiring addi-
ments and strategies to gain marketing approval
in the shortest possible timeframe. Ultimately,
tional studies (aiming to ensure clinical data are
relevant to the local population) or having sig-
3
the company may ask a regulatory professional nificantly longer approval times may negatively
the likelihood of achieving the product’s targeted
influence a country’s prioritization. Increased 4
labeling claims and when it will be achieved.
development costs or delayed market entry to
Increasingly, the overall goal is a global
development program with simultaneous, mul-
generate additional data may reduce financial
returns compared to marketing a product in a
5
timarket submissions and approvals. However,
many factors can influence the need for prioritiz- country without these challenges. Ideally, a com-
ing an individual country or region, identifying pany will consider an integrated evidence gener- 6
opportunities to enable early or more cost-ef- ation strategy that considers evidence required by
fective market entry. For example, orphan drug
programs vary significantly. Some are limited to a
the healthcare system’s multiple stakeholders and
will gather such evidence in the smallest number
7
reduction in regulatory agency fees, while others, of separate studies minimizing cost while meet-
like the US, have more generous programs that ing stakeholder objectives for evidence. 8
include additional agency support and devel- Clearly articulating claims and global market
opment grants. A second important example is
FDA’s accelerated approval program that pro-
entry timing enables potential sales in key mar-
kets, development costs and return on investment
9
vides an opportunity to gain early approval based
to be modeled.
on surrogate or intermediate endpoints, thus
reducing premarket development costs and time,
In early development stages, many 10
unknowns and assumptions need to be built
and leading to early market entry.
Similar programs to support innovative into the model. Scenario planning and sensi-
tivity analysis will take these uncertainties into
11
new medicines are available in Europe, includ-
ing accelerated assessment of medicines of account, which will reduce as development
major public health interest via the Centralised progresses and challenges are overcome, creating 12
Procedure, especially ones that are therapeutic an increasingly robust financial model to support
innovations. Accelerated assessment usually takes
150 evaluation days, rather than the standard 210
business planning.
During development, decisions made by
13
days. In addition, in 2016, a program to enable the multidisciplinary team will impact poten-
accelerated assessment of PRIority MEdicines tial business outcomes, the financial return and 14
(referred to as PRIME) was introduced. PRIME subsequent patient access. Decisions should
eligibility criteria are similar to those for acceler-
ated assessment but result in additional dialogue,
be based on data available, moving the prod-
uct forward toward the overall TPP objectives.
15
support and connectivity within the EU regu-
If available data indicate deviations from the
latory network. It is intended for medicines in
development to address an unmet medical need.
planned pathway are required, a detailed TPP 16
There also are mechanisms in some jurisdictions review and business outcome impact evaluation
to allow approval based on more limited data should follow.
The sponsor TPP and development plan are
17
(e.g., Phase 2 studies or surrogate endpoints)
due to the new medicine’s promising nature. The living documents and require regular monitoring
conditional approval mechanism in Europe is and review to evaluate progress against objectives 18
intended for this purpose, where benefit-risk is and updating as appropriate.

189
Chapter 15: Global Regulatory Strategy

Figure 15-2. Key Interactions Between Marketing and Regulatory

Basic Clinical Regulatory Product


Postapproval
Research Studies Approval Launch

Product Target Product Product labelling Promotion and Product


potential Profile branding extensions
Product
Comparators presentation Advertising
Outcomes Filing strategy Postapproval
research studies

Marketing medicines’ approval is replaced by a cumulative


process based on gradual accretion of data (the
During all product development stages, the mar- ‘evidence continuum’). New therapies receive a
keting/commercial team contributes to the global conditional license based on further testing to
development strategy. To present a product to its substantiate their safety and efficacy in larger
target market, three key elements must be inte- or different populations, thus providing early
grated: product attributes, marketing decisions access to promising therapies, particularly for
and commercialization activities. Each of these unmet needs. Over time, more data are expected
may be influenced by the regulatory strategy, to be generated in the post-license setting, and
label claims and product presentation. access to technology and digital health records
The integrated product strategy will incor- will allow an environment that provides greater
porate the overall regulatory and market strat- confidence to regulators and other stakeholders
egies seamlessly. Key marketing interactions than has been possible before.
influencing regulatory strategy and vice versa are Marketing will be able to promote and
shown in Figure 15-2. build a new brand on an incremental basis.
Each territory should contribute to and take However, the opportunity for early market
into consideration the global product strategy launch also carries the risk that not all products
when developing regulatory and marketing strat- will have the anticipated benefits, as subse-
egies for local markets. quent trials may show a small treatment effect
Global regulatory environment changes or unacceptable adverse events. This risk needs
and precedence will impact marketing strategies to be communicated to patients and health-
directly. International harmonization of regula- care providers clearly and systems developed
tory requirements is increasing the opportunity to monitor new medicines in the marketplace
for companies to file and launch new products more closely. License cancellation or restriction
simultaneously in multiple markets. Further, the risks and consequences should be incorporated
introduction of live (adaptive) licensing, based on in risk management planning.
a medicine’s early approval in a restricted setting
Reimbursement
due to positive data in the limited setting and
shifting data generation in broader settings to a While significant efforts have been directed
subsequent line extension, will impact the sales toward global harmonization of regulatory
and marketing process significantly. Instead of an requirements, reimbursement processes and sys-
all-or-nothing regulatory approval approach, new tems are country-specific, with little or no har-

190
Global Pharmaceutical and Biologics Regulatory Strategy, Second Edition

1
monization among jurisdictions, partly because Late-stage clinical trial design consider-
each country’s clinical, social and financial ations impacting HTA assessments include: 2
context can be so varied. However, several guid- • Active comparators—is a three-way
ing principles underpin many health technology
assessment (HTA) and reimbursement systems:
design incorporating a placebo and an
active comparator feasible? How will
3
• Is the new medicine effective in the the comparator be selected, as regional
population? standards of care may vary? 4
• Does it have advantages over currently • Are quality of life and validated instru-
available therapies? ments to evaluate patient preferences
• Will it contribute to an overall health- planned? 5
care savings (i.e., offset other costs)? • Does the trial use broader patient pop-
• Is it affordable? ulations?
6
The product value concept is incorporated in the Optimally, preapproval clinical trials will incor-
sponsor TPP, with contributions from health porate these data requirements. Increasingly, 7
outcomes, marketing and other integrated prod- postapproval studies are required. These may be
postapproval commitments as a condition of a
uct development team members.
Generally, a new product’s value and answers
live licensing program or a risk management 8
program required by a regulatory agency. In
to these questions are not forthcoming from a
addition, postapproval observational studies and
clinical development program focused solely
registries provide evidence to support reim- 9
on achieving regulatory approval. Regulatory
bursement submissions and continue to remove
requirements compared with HTA authority
requirements are shown in Table 15-1.
uncertainty in the postmarket setting. 10
On its own, the data package the regulatory Global Product Development
authority requires for marketing approval often
will be insufficient to meet HTA data require-
CMC (Chemistry, Manufacturing and 11
Controls)
ments. In countries where reimbursement
is pivotal to a product’s market success, data A CMC product development program focuses 12
required to support reimbursement must be on the drug substance and drug product’s formu-
incorporated into the product development plan
and regulatory strategy.
lation, process development and presentation as
well as the manufacturing facility. From a global
13
14
Table 15-1. Comparison of Regulatory and HTA Requirements

Regulators HTA Authorities 15


Evaluate efficacy (therapeutic effect) Evaluate effectiveness, how well a treatment works in the practice of

Homogenous data
medicine
Heterogeneous data
16
Risk/benefit Cost effectiveness
Comparator:
• placebo (FDA)
Comparator:
• current standard of practice
17
• active drug (EMA)
Focus is on the drug Focus is on population, cost and generalizing beyond data 18

191
Chapter 15: Global Regulatory Strategy

perspective, the CMC development program with a simple formulation, but later clinical
must consider many aspects, including develop- studies should be conducted with the product
ment phase and regional requirements. planned for marketing. If significant manufac-
A global CMC strategy should be inte- turing or formulation changes are made late in
grated and linked with the overall global development, bridging studies may be required to
product strategy to ensure suitably formulated confirm the changes have not altered the prod-
product is available to meet a particular devel- uct’s safety or efficacy profile.
opment stage’s requirements. Other clinical study CMC considerations
The three key factors influencing the CMC include requirements for a placebo product
development program are quality, time and cost. identical in appearance to the active product for
All these factors are related, and the chosen use in double-blind studies. Placebo development
pathway also will depend on the business’s risk may be challenging if the active drug substance
tolerance. For example, an organization initially has characteristics difficult to emulate with an
may choose to save time to get the first patient inactive substance, e.g., smell or taste.
for a given clinical study. However, in the end, Many countries are members of the Phar-
that decision may incur higher costs and addi- maceutical Inspection Convention and Phar-
tional time or even result in a product profile that maceutical Inspection Cooperation Scheme
is less acceptable to patients (e.g., short storage (PIC/S), which provides a GMP guide for
conditions due to lack of stability data). medicinal products. Annex 13 sets out require-
Initially, a non-GMP drug substance is ments for clinical trial product manufacturing
acceptable for nonclinical studies, provided the and labeling. This guide can assist the CMC
impurity profile does not increase a toxicity risk team in planning the clinical trial product man-
that might not occur at later development stages ufacturing program and core labels; however, the
when impurity limits are tighter. Frequently, only team always should be aware of the geographic
the drug substance is required for initial pharma- locations in which studies will be conducted and
cology and toxicology studies. However, formu- individual country- and study-specific require-
lation effects must be evaluated, and the drug ments. In some countries, e.g., India, patient
product may be required for nonclinical studies if labels and information are required to be in the
the product required a complex formulation due local language, meaning up to six local languages
to the drug substance’s physicochemical prop- must be accommodated. Clinical trial supply
erties, e.g., low solubility, membrane permea- availability is a key factor in study start-up
bility, stability or interactions with formulation times and should be coordinated with the
excipients. clinical development team. Batch records may
As the product moves into clinical stud- be required for study approval, but manufacture
ies, the CMC strategy must take clinical trial timing should ensure, as far as possible, sufficient
product requirements into account in countries stability data are available to avoid the need to
where studies are planned. In most countries, change batches during the study.
full GMP compliance is required for all clinical As the product development program moves
study phases. However, some countries, including closer to market, it is important the CMC team
Australia, allow use of a non-GMP product for a understands the regulatory filing strategy, desired
first-in-human study in healthy volunteers. product profile for patients and priority target
The drug product presentation also must be countries to ensure regulatory dossier CMC sec-
consistent with the clinical development strategy tions meet both the format and content require-
to ensure the dosage form and dose unit meet ments for each country. International Council
protocol requirements. Early in development, it on Harmonisation (ICH) guidelines assist and
is recognized clinical studies may be conducted enable manufacturers to develop a core CMC

192
Global Pharmaceutical and Biologics Regulatory Strategy, Second Edition

1
dossier, but many individual country requirements Clinical Development
exist for non-CTD format, stability studies across 2
temperature zones, labeling and packaging. In Increasingly, the clinical development pro-
gram’s goal should not only be global but,
addition, specific product type guidelines devel-
oped by many jurisdictions may require additional essentially, simultaneous. 3
or different product specifications. Simultaneous global clinical develop-
Marketing and reimbursement teams also ment can bring many benefits to the global
regulatory strategy:
4
may have country- or region-specific require-
ments that must be accommodated to support • broader regulatory oversight—
commercial success, e.g., a pack size equivalent simultaneous submission and regulatory 5
to one month’s supply is the maximum quantity agency review allow the authorities’
views and experiences to be included in
permitted to be dispensed or reimbursed on
each occasion. the clinical development program 6
• reduced drug availability lag—successful
Nonclinical simultaneous clinical development
should result in earlier innovative drug
7
As the nonclinical plan is developed, it must therapy availability to populations in
be integrated with both the clinical and CMC
programs. Clinical studies must be supported
new markets and, ultimately, a reduction 8
in drug lag from core countries’
by appropriate nonclinical studies. For example, marketing time
nonclinical toxicology studies should use the • ethnic factors—broader clinical 9
route of administration proposed in the clinical development enables a science-based
study, of the same or longer duration. Results of
nonclinical studies conducted prior to the first
approach to define intrinsic and
extrinsic factors and identify meaningful
10
Phase 1 study should provide information to ethnic differences (In addition,
guide clinical study starting dose selection. Simi- registration dossiers will include higher 11
larly, reproduction study timing generally will be ethnic diversity.)
determined by the timing of including women of
child-bearing potential in clinical studies. Global clinical development data should satisfy
12
In countries that have adopted ICH not only US and EU requirements, but also
guidelines, the nonclinical program required to address ethnic differences and satisfy the local 13
support clinical studies generally is harmonized. data requirements for marketing applications
However, even in ICH countries, differences in countries such as Japan, China, South Korea,
exist in long-term toxicity study duration to sup- Taiwan, Mexico and India. 14
port Phase 3 trials and marketing applications. Global programs have contributed to the
For example, ICH M3(R2) on the duration of geographic shift in selecting countries for clinical 15
repeat-dose toxicity studies provides different trials. Drivers for this shift also include access
recommendations for the EU and US. In the EU, to wider, often treatment-naive populations and
a six-month repeat-dose study in two species a reduction in development costs. Increasingly, 16
(rodent and nonrodent) is required. However, the countries outside the traditional drug develop-
US requires a nine-month nonrodent study. ment core countries are offering quality sites and 17
Therefore, the nonclinical plan must meet good investigators.
local regulatory requirements to initiate clinical Organizations may set out to design a global
studies and meet global nonclinical requirements clinical development program, but whether this 18
needed to support the market application. can be truly simultaneous depends on many

193
Chapter 15: Global Regulatory Strategy

factors within each country, including regulatory ization and identifying the steps and regulatory
requirements and start-up times. requirements necessary to achieve this may
The unpredictable regulatory environment can influence program design significantly. For
be a major deterrent to global clinical development: example, if in-country clinical trials are needed
• lack of harmonized requirements and for regulatory approval, it may be possible to
processes among countries, and unique plan the clinical development program to include
data requirements outside international these countries in global trials in addition to
standards in-country trials.
• variability in acceptability of novel Some countries may require specific local
approaches, e.g., innovative clinical nonclinical studies or clinical pharmacokinetic
study designs studies prior to starting later-phase studies.
• variable and inefficient review of initial A global Phase 3 program may include local
approval and amendment processes requirements for ethnicity or a specific percent-
• data acceptance and uncertain ICH age of patients from each region.
GCP enforcement can raise doubts Failure to determine these requirements
about whether trial results will be prior to starting the registration studies could
acceptable to other agencies result in significant cost implications and delays
• intellectual property concern barriers in marketing application submissions if they are
to providing detailed information (e.g., not met.
CMC) at an early stage Figure 15-3 provides an example of the reg-
ulatory strategy to reduce approval timelines in
When planning a global clinical development key Asian markets by including these countries
program, prioritizing markets for commercial- in a global clinical development program.

Figure 15-3. Clinical Development Strategy in Asia

Marketing Approval
(US/EU)

US/EU IND or CTA Phase 1 Phase 2 NDA Launch

Phase 3

Marketing Approval
Japan IND or CTA Phase 1 Phase 2 NDA

Taiwan NDA
TW: Meet DOH Briding
Korea Dose Finding Requirement
KR: Meet KFDA Briding NDA
China Requirement
CN: Meet China NDA
Requirement
Drug Lag
Source Country CPP 2–2.5 Years
CTA: Clinical Trial Approval
CPP: Certificate of a pharmaceutical product to start NDA
application “save 2–3 years”

194
Global Pharmaceutical and Biologics Regulatory Strategy, Second Edition

1
Clinical study protocol design also may Dossier
influence data acceptability in different jurisdic- Core Dossier
2
tions. Regulatory guidance on developing new
therapies for specific indications will influence
overall study design, including objectives, end-
Regulatory agencies expect an assurance of
medicines’ quality, safety and efficacy before they
3
points, use of active comparators or placebo and authorize their distribution to patients in their
study duration. In an effort to support conduct of countries. 4
clinical studies to meet requirements in both the While the information required by each
country’s regulators differs, ICH has developed
EU and US, EMA and FDA provide organiza-
tions the opportunity to seek parallel Scientific some harmonization of standards among coun- 5
tries. In addition to the three ICH regions (US,
Advice. The goal of parallel Scientific Advice is
sharing information and perspectives, rather than
Europe and Japan), many countries have adopted
ICH guidelines, including Australia, Switzerland
6
necessarily resulting in harmonization of regula-
and Canada.
tory requirements. However, for innovative new
products for which no specific guidance is avail-
ICH has developed a standard content for- 7
mat, the Common Technical Document (CTD),
able, agreement on development requirements that also is organized with consistent sections
may be a beneficial outcome. Increasingly, it and headings2 (see Figure 15-4). 8
also is desirable to obtain advice in parallel with Module 1 is for administrative information
reimbursement authorities to drive alignment
in what the regulatory authority and subsequent
and prescribing information and should con-
tain region-specific documents, e.g., application
9
reimbursement authorities would prefer to see in forms or the proposed regional label. Module 1
the overall evidence package. is outside the formal ICH guidelines, and each 10

Figure 15-4. The CTD Triangle


11
12
No
tp
art

13
of

Module 1
the

Regional
CT

Administrative
D

Information
14
CTD Table of Contents
2.1
CTD Table of Contents
15
2

2.2
le
du

Nonclinical Clinical
Mo

Quality Overview Overview


2.4 2.5
Overall 16
CT

Summary Nonclinical Written and Clinical


D

2.3 Tabulated Summaries Summary


2.6 2.7

Module 3 Module 4 Module 5 17


Quality Nonclinical Clinical Study
Study Reports Reports
18

195
Chapter 15: Global Regulatory Strategy

jurisdiction that has adopted ICH guidelines also regulatory reviewers to focus on the
has developed specific Module 1 guidance. product’s key outstanding issues
ICH publishes structure and content guide- • ensuring the messages delivered are
lines for Modules 2–5. simple, clear and succinct to aid the
Module 2 contains summaries of the organi- agency’s regulatory review and decrease
zation’s position on available data and summaries agency questions
of quality (QOS), safety (CSS, NCO) and effi-
cacy (CSE, CO). Regulators require the sponsor Organizations generate the filing documents
to provide its assessment of the product’s overall required to complete each CTD section, but
benefit-risk balance in this section, including any following initial approval, may need to modify
outstanding uncertainty in either risk or benefit specific sections using a variation, amendment or
at the time of initial regulatory review. supplement. For example, additional clinical data
Module 3 provides information related to may be generated with the intent of expanding
how the product was developed, how it is man- the product’s use, and the sponsor will be required
ufactured, evidence of product stability under to provide a new clinical study report (CSR) and
standard and stressed storage conditions and data proposed label changes to support the new use.
ensuring it can be manufactured reproducibly Companies often initially generate an ICH-
and analyzed to generate a reproducible product style dossier first and use this to generate the
meeting an appropriate release specification. dossiers for non-ICH countries.
Module 4 provides information related to
completed nonclinical analyses. Regional Dossiers
Module 5 provides the sponsor’s clinical trial
study reports demonstrating a tolerable safety The dossier’s content and structure may need to
profile and positive beneficial effect. be modified to meet each regulatory authority’s
ICH also has authored guidelines (quality, requirements in the region or country in which
safety and efficacy) clarifying expectations for the the organization wishes to register the product.
information these sections should contain. This may include a range of additional consider-
For a global organization, the goal is to gen- ations. Some examples are provided below, and
erate a core regulatory dossier efficiently that can specific requirements are called out at a high
be reused in multiple markets where it may wish level in Table 15-2.
to register the product. Generating this dossier, • Providing extra data—some regulatory
therefore, may take some regional differences and agencies require additional clinical
requirements across markets into account. For data representing the local population,
example, the core quality section may describe e.g., China, Japan, etc., or data with an
process information related to a range of man- alternative comparator representing that
ufacturing facilities supplying several markets, market’s standard of care.
allowing this information to be selected for the • Additional declarations—legal letters of
relevant local dossier. authority, Certificates of Analysis, exe-
A regulatory professional has multiple roles cuted batch manufacturing records all are
in generating the regulatory dossier: examples of country-specific additions.
• advising on content requirements • Local forms—application forms, local
• ensuring coordinated delivery of multi- language versions of clinical summa-
ple components ries, Certificates of Pharmaceutical
• ensuring a balanced benefit-risk Product (CPPs) confirming prior
assessment that acknowledges product approval in another jurisdiction (typi-
weaknesses and uncertainty, allowing cally US or EU).

196
Global Pharmaceutical and Biologics Regulatory Strategy, Second Edition

1
Table 15-2. Overview of Specific Country Requirements 2
Country Global
Dossier
Local Trials Local Language CPP
Required
Specific Requirement
3
US ICH CTD Representative ethnic mix English No Patient-level data
Integrated

Summary of Safety and 4


Efficacy
EU ICH CTD Representative ethnic mix English, plus local language No
required for product
information
5
Japan ICH CTD Required Japanese No
Australia ICH CTD Generally No, except
bioequivalence studies using
English No Australian-specific Risk
Management Plan
6
local product as comparator

7
China Local format Required Chinese (Mandarin) Yes Extensive dossier
required for clinical
trial approval
South Mixed—ICH Mixed—some countries English, plus local language Mixed
America CTD and
local format
require local data, some
do not
required for product
information 8
9
• Alternate formats—some regions, like requiring alternative storage or stability
the Association of Southeast Asian data.
Nations (ASEAN) require their own • Clinical differences—intention to 10
dossier format (e.g., the ASEAN register an alternative indication or
CTD). This format incorporates both
summaries and detailed reports into
additional local data. This data inclusion
may require a new clinical review or
11
one part, e.g., Part IV includes both modification of other sections, e.g., inte-
the clinical summary and clinical grating new data into safety summaries 12
study reports. Formatting at a practical and summary tables. Some markets also
level can be as specific as the color may require clinical data comparing
of the tabs between sections or page the product to that market’s current 13
numbering, and care must be taken to standard of care.
meet these formatting requirements for
countries retaining these specific dossier
• Timing difference—depending on when
the original filing is submitted, a cur-
14
validation needs. rent dossier version incorporating the
sequence of changes that have occurred 15
An alternative commercial strategy may may be required. For this purpose, good
practice is to keep a ‘current master ver-
require tailoring the dossier, e.g., where differ-
ent data are required to register attributes for a sion’ in addition to tracking versions and 16
specific market. content registered in each market.
• Quality differences—registering Planning Filing Sequence Across Global 17
alternative presentations, manufactur- Markets
ing sites or supply routes in a specific
country, different release specifications The dossier generation and filing sequence 18
or humidity and temperature conditions planning strategy is the regulatory professional’s

197
Chapter 15: Global Regulatory Strategy

responsibility, in consultation with the global (RMPs) to be submitted as part of marketing


multidisciplinary development team. Some applications and require formal postapproval
markets do not accept applications until one of activities to ensure appropriate prescribing and
the ICH (i.e., reference) countries completes usage and to monitor and manage product safety.
its review. Once this initial review is complete, Although detailed RMP guidelines and formats
a CPP can be issued confirming a satisfactory vary across jurisdictions, the risk management
review was completed in another jurisdiction. principles articulated in the International Stan-
This can mean other markets can do a slightly dard ISO 31000 generally apply.3
abbreviated review based on the fact the prior From early planning and as product devel-
review has occurred, known as a reliance mech- opment progresses, ISO 31000’s principles can be
anism. This allows regulatory submission to take applied to enable early product risk identification
place in waves. and assessment, together with mitigation plans to
Key considerations when planning a filing minimize these risks during further development,
timeline include: at product launch and postapproval.
• availability of required filing data, e.g., When planning and preparing marketing
local market data
applications for each jurisdiction, the residual
• prior reviews, e.g., in an ICH country
risks and management strategies identified in
(US, EU, Japan) (including an ability
a global RMP can be customized to produce
to leverage responses to these questions
jurisdiction-specific RMPs.
elsewhere)
• launch time commercial driver During the evaluation process, the regula-
(impacted by market dynamics), e.g., tory agency may identify new risks or request
when does the organization want to alternate management strategies. These may be
initiate distribution, as soon as possible country-specific issues, such as those associated
or at a defined time with prescribing patterns or healthcare practices.
• resource availability to complete the In these instances, country-specific responses are
filing (including responding to agency required. Alternatively, these may potentially be
questions) issues relevant around the globe. Coordinating
• desired filing sequence (knowing agen- global risk management planning and incorpo-
cies discuss reviews with each other, and rating these issues into RMPs and postapproval
agreement on specific label language activities provide the opportunity to maintain
in one region can impact the review a globally harmonized lifecycle maintenance
outcome in another) approach. Issues initially restricted to one country
Lifecycle Management or jurisdiction may, in time, be applicable to other
countries, and the understanding and knowledge
Postapproval Commitments gained then can be shared for mutual benefit.
At the time of initial registration, a company The need to globally coordinate postapproval
will have limited experience and exposure with commitments will increase as adaptive licenses
its drug and some uncertainty. In the past, are granted. Adaptive licenses acknowledge data
regulatory approval was ‘all or nothing.’ When a continue to accumulate after a license is granted,
product is approved for marketing, a regulatory and access is best addressed by repeat cycles of
agency has relied almost exclusively on sponta- learning and confirming (re-licensing) over the
neous adverse reporting programs to monitor a product lifecycle continuum.
new product’s safety in the marketplace. Increased monitoring of real-world perfor-
More recently, an increasing number of mance (Real World Evidence, RWE), including
jurisdictions require Risk Management Plans postauthorization efficacy and safety studies

198
Global Pharmaceutical and Biologics Regulatory Strategy, Second Edition

Figure 15-5. Global Technology System for Safety Monitoring 2


Clinical Data
System 3
Central
Clinical Trial Laboratory

4
Management System
Site Data Entry System

Global Safety
Signal
Detection
External
Regulatory
5
Incoming System Database Database
ADR

6
Reports
Literature and
Spontaneous ADR
Reports

7
Routine
Reporting

Significant

8
Alert
Case Case Series Signals Queries
Reports Regulators

Product Safety Monitoring


9

and registries, will be required. This is especially Safety Monitoring 10


true when smaller data packages were initially
relied upon to gain regulatory approval and this
evidence may be required by other stakeholders
Clinical product development risk assessment
must be thorough and rigorous. However, it is
11
such as reimbursement authorities. For countries impossible to identify all safety concerns during
with relatively small populations, global coop- controlled clinical trials. Once a product is mar- 12
eration among regulatory agencies and global keted, the number of patients exposed generally
studies are required to provide the breadth and increases dramatically, including those with
depth of additional data required to support co-morbid conditions and/or taking concomitant 13
ongoing approval. Overall, this lifecycle man- medications. Therefore, postmarketing safety data
agement approach is expected to lead to lower collection and clinical risk assessment are critical
for evaluating and characterizing a product’s risk
14
patient risks compared to the current approach,
despite smaller early data packages.4 profile and making informed risk minimization
The International Coalition of Medicines decisions. The growing trend for early approvals 15
Regulatory Authorities (ICMRA), formed in based on less-extensive clinical trial data further
increases this imperative.
December 2013, provides a forum for develop-
ing a cooperative approach among regulators on Postmarket safety data monitoring now 16
many topics, including postapproval commit- is a universal regulatory authority requirement
ments. This group and other such coalitions of for marketing approval and may include formal 17
authorities in multiple jurisdictions can utilize postapproval clinical studies, monitored release
reliance mechanisms, working together to share programs and spontaneous reporting programs.
reviews, guidelines and best practices, and can be Although these postapproval obligations are 18
useful tools when considering filing strategies. mandated by agencies at the jurisdiction level,

199
Chapter 15: Global Regulatory Strategy

reporting requirements are predicated on both drawn from the market many years after initial
local and global monitoring programs. launch due to long-term safety studies initiated
Safety scientists, therefore, must find ways after a series of spontaneous reports alerted regu-
to capture, record and analyze huge amounts lators to potential concerns.
of safety information across different studies, Opportunities exist for greater international
systems and jurisdictions and coordinate this collaboration among regulators who potentially
information globally to meet reporting require- have access to bigger pharmacovigilance datasets
ments, make informed recommendations and and data mining capabilities, as well as increased
implement decisions. collaboration and harmonization on regulatory
Global pharmacovigilance processes and decisions. The next wave in this workflow is the
workflow are supported most effectively by introduction of artificial intelligence to enhance
using a global pharmacovigilance system as data mining and uncover new insights from large
a technology tool for drug safety detections, data sets.
data mining, results interpretation and deci-
sion-making support. Such systems are trans- Maintenance and Compliance
forming industry’s capability to detect safety
Throughout the lifecycle, product registration
signals early as a key component of meeting
must be maintained in accordance with regula-
regulatory authorities’ postapproval require-
tions in each jurisdiction in which it is approved.
ments and demonstrating a commitment to
safety that transcends regulatory compliance. Variations to the current approved product may
An example of a global technology system require a submission and regulatory authority
supporting improved safety monitoring is shown approval, depending on the nature of the pro-
in Figure 15-5.5 posed change. Similarly, the approval timelines,
It is important to acknowledge a global and such provisions as grace periods (for imple-
technology system is a support tool and must mentation), vary widely in different regions.
be used within the global policy and procedure Whenever proposed changes are planned,
framework to establish a culture of safety and e.g., a quality change such as a packaging change,
quality and provide the expertise and resources at they ideally should be planned well in advance
both the global and local level. of implementation. A global plan is required,
Globally, these will include: including an understanding of region-specific
• establishing functional groups and requirements for the proposed variation, e.g.,
expertise data requirements, submission format, time-
• developing and aligning integrated lines and costs. It is good practice to assess the
operational procedures change’s intended impact and benefit, e.g., a
• developing and implementing well-de- minor change in manufacturing may take more
fined decision-making models, esca- time and effort to introduce than the savings in
lation processes and communication efficiency intended locally in the plant.
channels Similarly, changes relating to new clinical
• incorporating continuous improvement data availability to extend the approved indica-
activities tions may be planned well ahead of time at both
the global and local levels.
These principles apply regardless of the organiza- As with new product submissions, a spon-
tion’s size, the number of products or number of sor may elect to compile a core global variation
jurisdictions. Newly launched products generally dossier, accessible via a central database, with
are the focus of key postapproval monitoring. local requirements added at a regional or country
There are notable examples of products with- level. The submission then is managed at the

200
Global Pharmaceutical and Biologics Regulatory Strategy, Second Edition

1
local level, with local regulatory staff responsible • creating customized reports interpreting
for regulatory agency interactions. regulations and ensuring the regulatory 2
Other changes, such as safety-related updates function is the ultimate authority on all
following unexpected serious adverse events and
signals detected through postmarketing surveil- •
regulatory compliance aspects
tracking regulatory key performance
3
lance, often are accompanied by tight regulatory indicators with a balance of lagging
timelines for notifying the regulatory agency and
updating product information documentation to
indicators (e.g., issues generated after 4
product launch) and leading indicators
ensure new risks are communicated promptly to (e.g., revenue at risk from potential
patients and healthcare professionals. Although it
is not possible to plan specific regulatory interac-
regulation changes) 5
tions in advance, global planning for such events is
vital to enable rapid implementation if these occur.
Regulatory functions within organizations,
especially those with global programs, should not
6
A multidisciplinary team including experts in reg-
be merely reactive and tactical, but more proac-
ulatory and safety, compliance, risk management,
communications and marketing should document tive and aligned with the organization’s global 7
procedures and ensure all key staff (and backups) product strategy to ensure minimal regulatory
are trained and ready to implement the plan, compliance challenges and timely mitigation of
if necessary. A local plan should be developed, nonconformance risks. 8
consistent with each region and country, that links
Regulation Database
into the global plan.
9
Companies need to have reliable, current
Knowledge Management
knowledge databases to track technical and legal
Knowledge management is one of the most requirements for each country in which they 10
significant regulatory challenges. Regulatory intend to submit regulatory dossiers. Commercial
expertise often is comprised of knowledge of
what is written in regulation, knowledge of
databases are available, and/or companies can
utilize staff to provide local intelligence. Local
11
precedence and actual personal experience and direct intelligence advantages include supple-
a set of relationships with the relevant authority menting local published guidance with practical 12
that help the regulatory professional collectively insights and experience of what actually was
provide advice on what is ‘required’ and what is required in addition to what was published. If
‘expected’ to deliver a positive outcome. using a commercial database, the information’s 13
currency should be confirmed, as regulations
Experience Capture change frequently, especially in countries with
evolving regulatory frameworks.
14
Properly captured knowledge and centralized
information sources yield process efficiencies. Knowing the Regulatory Authority
Actions taken by companies for proper knowl- 15
edge codification and dissemination include: Organizations need to know not only the written
• creating an “expert tracker” database for
knowledge residing with the organiza-
regulatory requirements, they also should have a
good understanding of unwritten requirements
16
tion’s employees and attempting to cen- and the personal preferences of any agency
tralize and disseminate that information reviewers who may review their dossiers. 17
• disseminating information through If possible, regulatory personnel should be
multi-level training at different career encouraged or advised to interact with the regu-
stages for regulatory and affiliated lators during new product development, although 18
personnel it is not possible or efficient to meet with all

201
Chapter 15: Global Regulatory Strategy

agencies. These interactions can help the orga- Therapeutic Goods Administration or others. Key
nization get to know the future agency review agency meeting considerations may include:
team, help that team become familiar with the • data reporting impacting benefit-risk
product’s innovative aspects and gain input on conclusion and requiring review before
the proposed strategy. advancing to the next phase
All regulatory agencies expect companies • clinical design issues requiring discussion
to be truthful, transparent, collaborative, sci- (e.g., where there is an intention
ence-based and patient-focused in all interactions. not to follow regional guidance and
expectations or where a specific
Language, Culture and Local Insights innovation is intended in the trial design)
To be successful, it is important companies • defining milestone meetings, e.g., End-
understand which documents must be presented in of-Phase 2 (EoP2)
the local language. This might include packaging, • technical reviews with specific
patient or physician leaflets, clinical summaries, etc. committees, e.g., biotechnology
Holding meetings in the local language can products or advanced therapies
show respect for the culture, especially where facil- • preapproval reviews, e.g., Advisory
itated by a representative from the country who is Committee Meeting
aware and respectful of specific expectations. For • presubmission interactions to familiarize
global companies, this step can be cumbersome. the review team with the dossier
Local language negotiations or discussion and allow in-person discussion and
of highly scientific dossier review aspects also clarification ahead of dossier submission
may be more fruitful in the local language
if the reviewers are less comfortable with an The global product strategy team’s job is to
alternative language. define the overall corporate product development
Some countries have specific format require- strategy, assimilate the feedback from multiple
ments, which may extend to electronic format, jurisdictions (and possible stakeholders if joint
paper color for the printed dossier, signatures and HTA advice is also sought) and propose a plan
required dossier page numbering, etc. Insights that takes feedback obtained into account. It is
into each of these requirements will ensure good practice to document the advice received
speedy validation and subsequent dossier review. and capture why the company chose not to
follow that advice, as this is likely to come up
Regulatory Authority Meetings during the subsequent marketing authorization
application review.
Global organizations may plan to interact with
specific regulatory agencies to gain feedback on Electronic Submissions
available data and intended next development
process steps. In general, companies will interact To facilitate submission, navigation and review,
with only a limited number of regulators to get many countries accept electronic dossiers. To
a sense of their intended action’s likely accept- ensure presentation is in a common format,
ability. Companies intending to market in the ICH has agreed on a common format known as
US normally will interact with FDA at defined the electronic Common Technical Document
milestones; similarly, if intending to market (eCTD).6 The table of contents is consistent
elsewhere, key interactions may include meetings with the CTD, and the document is structured
with EMA (and/or specific national European in HTML format. Since this field is evolving,
regulatory authorities), Japan’s Ministry of Health, current standards should be verified via ICH or
Labour and Welfare, Health Canada, Australia’s country-specific websites prior to dossier com-

202
Global Pharmaceutical and Biologics Regulatory Strategy, Second Edition

1
pilation and submission. For example, since July References
2015, eCTD submissions have been mandatory 1. Draft Guidance for Industry and Review Staff: Target 2
for all EMA centralized applications. Product Profile—A Strategic Development Process Tool.
Many countries do not accept eCTD yet, but Draft (2007). FDA website. https://www.fda.gov/
may accept a non-eCTD electronic submission
media/72566/download. Accessed 10 October 2019. 3
(NEES). NEES differs from an eCTD because it 2. Organisation of the Common Technical Document for the
Registration of Pharmaceuticals for Human Use M4(R3).
does not have an XML backbone or MD5 check-
sum, as defined by ICH. Rather, it is a collection
ICH website. https://database.ich.org/sites/default/
files/M4_R4__Guideline.pdf. Accessed 26 November
4
of electronic files organized in folders. 2019.

International Cooperation and


3. International Organization for Standardization.
ISO 31000: 2009 Risk management Principles and
5
Harmonization Guidelines.

As organizations seek to increase their drug


4. Eichler HG, Baird LG, Barker R, Bloechl-Daum B,
Borlum-Kristensen F et al. “From adaptive licensing
6
development programs’ efficiency by increasing to Adaptive Pathways: delivering a flexible life span

7
approach to bring new drugs to patients.” Clin
their focus on early and simultaneous registra- Pharmacol Ther 2015; 97: 234–246.
tion in multiple global markets, regulators also
5. Lu Z. “Information technology in pharmacovigilance:
are growing more aware of the need for global
8
benefits, challenges and future directions from industry
harmonization of requirements and processes. perspectives.” Journal of Drug Healthcare and Patient
Efforts to increase global harmonization are con- Safety. 2009:1 35–45.

tinuing through a range of activities in addition


9
6. Electronic Common Technical Document (eCTD) M8.
ICH website. https://www.ich.org/page/ich-electronic-
to ongoing ICH processes.
common-technical-document-ectd-v40. Accessed 26
November 2019.
ICMRA
10
ICMRA provides direction for a range of areas
common to many regulatory authorities’ missions.
It identifies areas for potential synergies among
11
regulators and facilitates, where possible, interna-
tional leveraging and resource savings by building 12
confidence and deeper regulator collaboration.

International Generic Drug Regulators 13


Programme (IGDRP)
IGDRP was created to promote collaboration and 14
convergence in generic drug regulatory programs
for regulatory agencies from several countries.
15
International Regulators Consortium

This consortium was formed in 2007 by like- 16


minded regulatory authorities to promote greater
regulatory collaboration and alignment of regu-
latory requirements. 17
18

203

You might also like