Sunteți pe pagina 1din 5

Changes

to an
Approved
Product
Chemistry, Manufacturing
and Controls
By Khandan Baradaran, PhD and Peggy Berry, MBA, RAC

I
t is a huge achievement for any company be foreseen, legitimate needs to make changes
to obtain licensing rights to an approved to the approved product or production process.
pharmaceutical product. Postapproval Postapproval changes can be relatively minor,
challenges are inherent in the marketing such as like-for-like changes to manufacturing
and distribution process. Of these, foreseen and equipment; or they can be more critical, such as
unforeseen changes to the manufacturing process a change to the drug product container closure.
top the list. Sponsors of newly licensed products The latter is common for products that undergo
may encounter issues that were not fully realized lifecycle management. For example, the compa-
during development, such as long-term stability ny may, develop and obtain approval for the drug
issues, which only come to light within the first supplied in a vial, then plan to obtain approval
two years postapproval. Such unforeseen issues for a prefilled syringe presentation for ease of
require corrective actions that usually impact patient use.
the approved manufacturing process in one or Certain changes must be reported to regu-
more ways. At the same time, there are likely to latory authorities, and in some instances, the

30 April 2008
sponsor may be prevented from distributing the regulatory agency needs to approve the change
drug manufactured with a particular change before distribution. This can be a complex task,
until the regulatory authority has reviewed and involving extensive oversight, reporting and
approved the change. In the US, manufactur- control within a company to identify a change,
ing changes can be reported to FDA in supple- assess the impact to the drug product, tag out
ments to the marketing/license application and the impacted drug product lots until approval is
in annual reports. The four reporting categories obtained, and report the change to the relevant
are: prior-approval supplement (PAS), changes authorities. The complexity increases with more
being effected (CBE), changes being effected in products in the manufacturing plant that may
30 days (CBE-30) and annual report (AR) (Table require changes, and marketing rights in mul-
1). Some changes are transparent to the license1 tiple countries.
and do not need to be reported. It is the regula- In the US, licensed pharmaceutical drug
tory professionals job to determine whether a manufacturers pay a postmarketing user fee to
change needs to be reported, and whether the market the drug (see Prescription Drug User Fee

Regulatory Focus 31
Table 1. Impact Assessment and Reporting of CMC Changes
Degree of Change Impact Assessment Reporting Category
Major change substantial potential to have an adverse effect prior-approval supplement (PAS)
Moderate change moderate potential to have an adverse event changes being effected in 30 days (CBE-30)
moderate potential to have an adverse event and
Certain moderate changes FDA concurrence obtained for product distribution changes being effected (CBE or CBE-0)
when supplement received
Minor change minimal potential to have an adverse event annual report

Act (PDUFA)).2 Fees are collected from licensed trols, equipment or facilities established in
pharmaceutical companies on a per-product (each the approved license application.3,4 To assess
approved dosage form) basis. Annual establish- a change, the regulatory professional must
ment fees and fees required for supplements with determine what potential adverse effects it
clinical information also apply. In return, FDA is could have on the products identity, strength,
required to review certain supplements within a quality, purity or potency as related to the
specified time frame (four months for most PAS, products safety or effectiveness. Which of the
30 days for CBE-30); some supplements have no four reporting categories applies depends upon
time constraints. this product impact assessment.
In addition to determining which submis-
Impact Assessment sion type to file (based upon potential effect on
Each change must be assessed to determine product quality attributes), the impact assess-
whether it needs to be reported. Changes ment must include any repercussions for vali-
that must be reported include changes to the dation; equipment, the manufacturing process
product, production process, quality con- step, analytical methods or any combination

Table 2. Major Changes (Substantial Potential of AE) Requiring Prior-approval Supplement Filing
change to validated sterilization process (sterile products)
Process Changes new/revised recovery procedures
new/revised purification process, including column change
change in process solution chemistry or formulation
change in processing step sequence or processing step addition, deletion or substitution
reprocessing a product without a previously approved protocol
scale-up requiring larger fermenter, bioreactor or purification equipment (biologics)

replacing an existing analytical method with a new one (e.g., RP-HPLC to replace ELISA)
Changes to establishing wider specification limits
Analytical Methods deleting a specification or analytical method
eliminating tests from the stability protocol
assay transfer to another QC laboratory
change in product composition or dosage form or ancillary components
expiration dating period extension or change in storage temperature
Other Major Changes container/closure change
change in manufacturing site
conversion from single- to multi-product manufacturing area(s)
culture growth time extension resulting in significant increase in cell doublings beyond validated
parameters (biologics)
cell line change or new MCB (biologics)
Triggers for Additional formulation or excipient changes
Clinical or multiple simultaneous changes (e.g., scale-up, media components impacting titer, change in
purification resin type, container closure)
Nonclinical Studies
any change resulting in an observed change in activity assay (e.g., out-of-specification or trend
regarding licensed specifications and manufacturing history)
significant impurity profile increase

32 April 2008
Table 3. Moderate Change Examples (moderate potential of AE) Requiring CBE Supplement Filing
addition of duplicated process change (e.g., fermentation) or unit process (e.g., purification column)
Changes Being with no change in process parameters
Effected in 30 Days (CBE-30)
manufacture of an additional product in previously approved multiple-product manufacturing area
with same equipment/personnelif no changes in validated cleaning and changeover procedures
additional release tests and/or tightening of specifications
Changes Being Effected (CBE-0) changing from one compendial test method to another
replacing an in-house reference standard

of quality systems may need to be revalidated. assessment as well as discussions with FDA in
Validation data may need to be included in the advance of the submission to obtain concur-
submission package with the data that demon- rence on the filing strategy and timing of the
strate comparability before and after the change. change (Table 3). Some examples are the addi-
tion of a duplicate process chain or operation
Submissions unit that does not impact licensed/approved
In the US, the waiting period when reporting process parameters, the addition of new prod-
can be reduced if a protocol for assessing and ucts in the licensed manufacturing areas, chang-
determining comparability is submitted and es to compendial test methods and the replace-
approved prior to any changes being made. ment of reference standards. CBE reporting is a
The comparability protocol should describe the rather gray area, and the reporting category may
change or possible changes, planned testing and change depending upon the availability (or lack
revalidation studies and conformance to prespeci- thereof ) of agreements with FDA and internal
fied acceptance criteria. This, in effect, allows a strategic needs.
sponsor to obtain FDA agreement on what stud- Changes with the least potential for
ies are expected when certain changes are made, impacting product quality attributes should
and acceptable comparability outcomes. be submitted in the Annual Report (Table
A comparability protocol should be filed 4). These can include additional or more-
with the initial marketing application or as a stringent release or stability specifications,
PAS. It allows FDA and the sponsor to agree on more-stringent stability test parameters or
methods to be used to demonstrate compara- time points, and modifications to analytical
bility between the old and new manufacturing test procedures that do not change the basic
process, including: test methodology or acceptance criteria. Drug
analytical methods and acceptance substance and drug product stability trends and
criteria data are also included in the AR.
stability data requirements5
statistical tools Summary
nonclinical studies, if needed Postapproval changes to manufacturing pro-
clinical studies, if needed cesses and quality controls present unique chal-
lenges to licensed drug companies and must be
Table 2 gives examples of major changes. assessed for their potential impact. The regula-
Significant process changes, formulation tory professional plays a pivotal role in assessing
changes, widening acceptance criteria, replacing the change, obtaining feedback from regulatory
analytical methods, scale-up and container authorities, and acting as the liaison between
closure changes typically require PAS. Triggers
for additional clinical or nonclinical studies
may include formulation changes or new Table 4. Minor Change Examples (Minimal Potential for AEs)
excipients, significant changes to activity or to File in Annual Report
impurity levels and, for biologics, cell line modification in analytical procedures with no change in basic
changes. Regulators are especially concerned Annually test methodology or release specifications
about the potential immunogenicity impact of reportable additional release tests and/or tightening of specifications
formulation or cell line changes for biologics. replacing an in-house reference standard
Certain moderate changes can be reported more-stringent stability test parameters or time points
as CBEs. Sponsors may submit a CBE-0 or report of drug substance and drug product trend analyses
CBE-30, depending in part upon the impact (release and stability data)

Regulatory Focus 33
the regulatory agencies and the in-house quality,
manufacturing and supply chain groups. F

REFERENCES
1. Sufficiently described in the license, such as a like-for-
like equipment change.
2. For more information and to see the updated PDUFA
2007. Accessed 4 January 2008.
www.fda.gov/oc/initiatives/advance/fdaaa.html.
3. Labeling change reporting requirements are beyond the
scope of this article and are not discussed herein.
4. 21 CFR 314.70 and 21 CFR 601.12.
5. Stability data may be required for both API/drug sub-
stance and final drug product, depending upon the
potential impact of the change to either.

Additional Bibliographic Material


1. International Conference on Harmonization (ICH)
Q5E: Comparability of Biotechnological Products
Subject to Changes in their Manufacturing Process
November 2004.
2. 21 CFR 314.70 (New Drug)
3. 21 CFR 601.12 (Biologic)
4. Changes to an Approved NDA or ANDA. FDA CBER/
CDER, April 2004.
5. Changes to an Approved Application for Specified
Biotechnology and Specified Synthetic Biological Products.
FDA CBER/CDER, July 1997.
6. Comparability Protocols Protein Drug Products
and Biological Products. FDA CBER/CDER, Draft
September 2003.
7. Demonstration of Comparability of Human Biological
Products, Including Therapeutic Biotechnology Derived
Products. FDA CBER/CDER, April 1996.

AUTHORS
Khandan Baradaran, PhD, is a Senior Manager of
Regulatory Affairs at Dyax Corp. Prior to joining Dyax,
she held management positions in analytical development,
quality control, quality assurance and regulatory affairs at
Biogen Idec.
Peggy J. Berry, MBA, RAC, is Senior Vice President,
Quality and Regulatory Affairs at Dyax Corp., responsible
for the strategic management and oversight of the quality
and regulatory affairs departments. Previously she was
director of regulatory affairs at AstraZeneca and managed
the regulatory and clinical departments at Dey Laboratories.
Berry also has worked for ILEX Oncology and Cato
Research Ltd.) and in FDA review divisions.

34 April 2008

S-ar putea să vă placă și